CA2191776A1 - Automated tissue assay using standardized chemicals and packages - Google Patents

Automated tissue assay using standardized chemicals and packages

Info

Publication number
CA2191776A1
CA2191776A1 CA002191776A CA2191776A CA2191776A1 CA 2191776 A1 CA2191776 A1 CA 2191776A1 CA 002191776 A CA002191776 A CA 002191776A CA 2191776 A CA2191776 A CA 2191776A CA 2191776 A1 CA2191776 A1 CA 2191776A1
Authority
CA
Canada
Prior art keywords
sample
receptacle
workstation
disposed
substance
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA002191776A
Other languages
French (fr)
Inventor
Steven A. Bernstein
Page A. Erickson
Stephen Barker
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Biotek Solutions Inc
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=22955357&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=CA2191776(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Individual filed Critical Individual
Publication of CA2191776A1 publication Critical patent/CA2191776A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • BPERFORMING OPERATIONS; TRANSPORTING
    • B25HAND TOOLS; PORTABLE POWER-DRIVEN TOOLS; MANIPULATORS
    • B25JMANIPULATORS; CHAMBERS PROVIDED WITH MANIPULATION DEVICES
    • B25J9/00Programme-controlled manipulators
    • B25J9/16Programme controls
    • B25J9/1679Programme controls characterised by the tasks executed
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B25HAND TOOLS; PORTABLE POWER-DRIVEN TOOLS; MANIPULATORS
    • B25JMANIPULATORS; CHAMBERS PROVIDED WITH MANIPULATION DEVICES
    • B25J9/00Programme-controlled manipulators
    • B25J9/16Programme controls
    • B25J9/1656Programme controls characterised by programming, planning systems for manipulators
    • B25J9/1671Programme controls characterised by programming, planning systems for manipulators characterised by simulation, either to verify existing program or to create and verify new program, CAD/CAM oriented, graphic oriented programming systems
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N1/00Sampling; Preparing specimens for investigation
    • G01N1/28Preparing specimens for investigation including physical details of (bio-)chemical methods covered elsewhere, e.g. G01N33/50, C12Q
    • G01N1/30Staining; Impregnating ; Fixation; Dehydration; Multistep processes for preparing samples of tissue, cell or nucleic acid material and the like for analysis
    • G01N1/31Apparatus therefor
    • G01N1/312Apparatus therefor for samples mounted on planar substrates
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N35/00Automatic analysis not limited to methods or materials provided for in any single one of groups G01N1/00 - G01N33/00; Handling materials therefor
    • G01N35/00584Control arrangements for automatic analysers
    • G01N35/0092Scheduling
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N35/00Automatic analysis not limited to methods or materials provided for in any single one of groups G01N1/00 - G01N33/00; Handling materials therefor
    • G01N35/0099Automatic analysis not limited to methods or materials provided for in any single one of groups G01N1/00 - G01N33/00; Handling materials therefor comprising robots or similar manipulators
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N35/00Automatic analysis not limited to methods or materials provided for in any single one of groups G01N1/00 - G01N33/00; Handling materials therefor
    • G01N35/00584Control arrangements for automatic analysers
    • G01N35/0092Scheduling
    • G01N2035/0093Scheduling random access not determined by physical position
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N35/00Automatic analysis not limited to methods or materials provided for in any single one of groups G01N1/00 - G01N33/00; Handling materials therefor
    • G01N35/10Devices for transferring samples or any liquids to, in, or from, the analysis apparatus, e.g. suction devices, injection devices
    • G01N35/1081Devices for transferring samples or any liquids to, in, or from, the analysis apparatus, e.g. suction devices, injection devices characterised by the means for relatively moving the transfer device and the containers in an horizontal plane
    • G01N35/109Devices for transferring samples or any liquids to, in, or from, the analysis apparatus, e.g. suction devices, injection devices characterised by the means for relatively moving the transfer device and the containers in an horizontal plane with two horizontal degrees of freedom
    • G01N2035/1093Cylindrical, e.g. variable radius and angle
    • GPHYSICS
    • G05CONTROLLING; REGULATING
    • G05BCONTROL OR REGULATING SYSTEMS IN GENERAL; FUNCTIONAL ELEMENTS OF SUCH SYSTEMS; MONITORING OR TESTING ARRANGEMENTS FOR SUCH SYSTEMS OR ELEMENTS
    • G05B2219/00Program-control systems
    • G05B2219/30Nc systems
    • G05B2219/40Robotics, robotics mapping to robotics vision
    • G05B2219/40097Select stations with mouse to create process steps
    • GPHYSICS
    • G05CONTROLLING; REGULATING
    • G05BCONTROL OR REGULATING SYSTEMS IN GENERAL; FUNCTIONAL ELEMENTS OF SUCH SYSTEMS; MONITORING OR TESTING ARRANGEMENTS FOR SUCH SYSTEMS OR ELEMENTS
    • G05B2219/00Program-control systems
    • G05B2219/30Nc systems
    • G05B2219/40Robotics, robotics mapping to robotics vision
    • G05B2219/40098Show grid locations with symbols of workstations
    • GPHYSICS
    • G05CONTROLLING; REGULATING
    • G05BCONTROL OR REGULATING SYSTEMS IN GENERAL; FUNCTIONAL ELEMENTS OF SUCH SYSTEMS; MONITORING OR TESTING ARRANGEMENTS FOR SUCH SYSTEMS OR ELEMENTS
    • G05B2219/00Program-control systems
    • G05B2219/30Nc systems
    • G05B2219/40Robotics, robotics mapping to robotics vision
    • G05B2219/40099Graphical user interface for robotics, visual robot user interface
    • GPHYSICS
    • G05CONTROLLING; REGULATING
    • G05BCONTROL OR REGULATING SYSTEMS IN GENERAL; FUNCTIONAL ELEMENTS OF SUCH SYSTEMS; MONITORING OR TESTING ARRANGEMENTS FOR SUCH SYSTEMS OR ELEMENTS
    • G05B2219/00Program-control systems
    • G05B2219/30Nc systems
    • G05B2219/40Robotics, robotics mapping to robotics vision
    • G05B2219/40108Generating possible sequence of steps as function of timing and conflicts
    • GPHYSICS
    • G05CONTROLLING; REGULATING
    • G05BCONTROL OR REGULATING SYSTEMS IN GENERAL; FUNCTIONAL ELEMENTS OF SUCH SYSTEMS; MONITORING OR TESTING ARRANGEMENTS FOR SUCH SYSTEMS OR ELEMENTS
    • G05B2219/00Program-control systems
    • G05B2219/30Nc systems
    • G05B2219/40Robotics, robotics mapping to robotics vision
    • G05B2219/40113Task planning
    • GPHYSICS
    • G05CONTROLLING; REGULATING
    • G05BCONTROL OR REGULATING SYSTEMS IN GENERAL; FUNCTIONAL ELEMENTS OF SUCH SYSTEMS; MONITORING OR TESTING ARRANGEMENTS FOR SUCH SYSTEMS OR ELEMENTS
    • G05B2219/00Program-control systems
    • G05B2219/30Nc systems
    • G05B2219/40Robotics, robotics mapping to robotics vision
    • G05B2219/40463Shortest distance in time, or metric, time optimal
    • GPHYSICS
    • G05CONTROLLING; REGULATING
    • G05BCONTROL OR REGULATING SYSTEMS IN GENERAL; FUNCTIONAL ELEMENTS OF SUCH SYSTEMS; MONITORING OR TESTING ARRANGEMENTS FOR SUCH SYSTEMS OR ELEMENTS
    • G05B2219/00Program-control systems
    • G05B2219/30Nc systems
    • G05B2219/45Nc applications
    • G05B2219/45092Analysing or chemical synthesis robot, moving samples from station to station
    • GPHYSICS
    • G05CONTROLLING; REGULATING
    • G05BCONTROL OR REGULATING SYSTEMS IN GENERAL; FUNCTIONAL ELEMENTS OF SUCH SYSTEMS; MONITORING OR TESTING ARRANGEMENTS FOR SUCH SYSTEMS OR ELEMENTS
    • G05B2219/00Program-control systems
    • G05B2219/30Nc systems
    • G05B2219/45Nc applications
    • G05B2219/45118Endoscopic, laparoscopic manipulator
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10TTECHNICAL SUBJECTS COVERED BY FORMER US CLASSIFICATION
    • Y10T436/00Chemistry: analytical and immunological testing
    • Y10T436/11Automated chemical analysis
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10TTECHNICAL SUBJECTS COVERED BY FORMER US CLASSIFICATION
    • Y10T436/00Chemistry: analytical and immunological testing
    • Y10T436/11Automated chemical analysis
    • Y10T436/115831Condition or time responsive

Abstract

A system which performs a plurality of independent analysis procedures simultaneously, possibly involving differing types of tissues and differing process steps, comprising a robotic arm (10), which may move the different tissue samples among a plurality of process stations (13), and a processor (15), which may select the next tissue sample to move, when to move it, and where to move it to. The processor may direct the robotic arm to interleave the differing process steps, adjust the process steps for conflicts, and optimize the order in which samples are moved. The processing stations may be disposed in a set of grid locations (12). The processing stations may comprise workstations for performing individual steps of the tissue assay procedures, such as solution trays. The processor may comprise a graphic interface by which an operator may specify the process steps, may monitor the progress of ongoing procedures, or override the determination of what process steps to perform.

Description

WO9.~l33~40 2 ~ 9 1 7 7 ~ 6 .

1 This application is submitted in the name of i~v~L~
2 8teven A. Bernstein, a citizen of the United States having a 3 postal address at 2717 San Marcos Avenue, Los Olivos, California 4 93441, and Page A. Erickson, a citizen of the United States having a postal address at 2505 Calle Montilla, Santa Barbara, 6 California, and 8tephen Barker, a citizen of the United States 7 having a postal address at 120-B Cremona Drive, Santa Barbara, 8 California 93109, all assignors to Biotek 801ution~, Inc., a 9 California cuLyur~Lion having an office at 120-B Cremona Drive, Santa Barbara, California 93109.

12S P _ _ I F I C _ ~ I O _ 15TITE~ OE THE IW ENTION

17 AUTOMATED TISSUE ASSAY USING STANDARDIZED CHEMICALS AND PAC-~AGES

19CROSS-REFF~FNCE TO RELATED APPLICATION

21This application is a continuation-in-part of 22 Application Serial No. 07/740,285 filed August 5, 1991, and 23 Application Serial No. 08~218,143, filed March 24, 1994, both 24 filed in the name of inventors Steven A. Bernstein and Page A.
Erickson, titled "Method and Apparatus for Automated Tissue 26 Assay", and assigned to the same assignee.

wog5133240 2 1 9 1 7 7 6 i~ 6 1 BACKGROUND 0~ THE INVENTIoN

3 1. Field of the Invention This invention relates to methods and apparatus usefu}
6 in automated analysis or testing of tissue samplesl and to 7 automated tissue assay using standardized chemicals and pa~k~C.

9 2. Pescrirtion of Related Art 11 The analysis of tissue is a valuable diagnostic tool 12 used by the pathologist to diagnose many illnesses and by the 13 medical researcher to obtain information about a cell structure.

In order to obtain information from a tissue sample it 16 is usually no~C~ry to per~orm a number of prsliminary 17 operations to prepare the sample for analysis. There are many 18 variations of the pLoceduL~ to prepare tissue samples for 19 testing. These variations may be considered refir ~ to adapt the process for individual tissues or because a particular 21 terhn;~l~ is better suited to identify a specific chemical 22 substance or enzyme within the tissue sample. However the basic 23 preparation techniques are essentially the same.

Typically such operations might include the processing 26 of the tissue by fixation, dehydration, infiltration and 27 ~ ; n~; mounting of the tissue on a slide and then staining 28 the sample; l~h~l;ng Of the tissue through the detection of WO95/33240 - 2 J ;91 776 r~ )............................ 156 , 1 various constituents; grid staining of tissue sections for 2 analysis by an electron microscope or the growing of sample cells 3 in culture dishes.

DPpPn~ing on the analysis or testing to be done, a 6 sample may have to undergo a number of preliminary steps or 7 treatments or pLoceduLes before it is ready to be analyzed for 8 its informational content. Typically the procedures are complex 9 and time c~nCllmi ng, involving many tightly sequenced steps often utilizing expensive and toxic materials.

12 These procedures must usually be performed in a 13 critical order for each sample and each treatment is frequently 14 time ~PpPn~Pnt. Additionally the lahoratory is often under extreme ~L~US~ to perform many different analysis as soon as 16 possible, entailing many different pLoceduLes and tests.

18 A sample of tissue may undergo an optical microscopic 19 examination 50 that the relationchip of various cells to each other may be detorminP~ or abnormalities may be u.l~uv~d. The 21 tissue sample must be an e~L~ ly thin strip of tissue so that 22 light may be transmitted th~lethL~h~l.. The average thickness of 23 the tissue sample or slice (often referred to as sections) is on 24 the order of 2 to 8 microns. A relatively soft and pliable tissue such as might come from an organ of the human body, in its 26 fresh state cannot be accurately cut into such thin sections. In 27 addition, in order to see the individual constituents of the 28 cells, such as the nucleus, the nucleolus, the cytoplasm and the ~o~.~l33240 2 1 9 :1 7 7 6 ~ r~

1 cell membrane, it is preferable to have them colored by different 2 dyes to produce a cffntrasting appearance between the ol ~ -n~C .
3 Very limited dye staining can be done on fresh or recently living 4 tissue without resorting to ~ho~ic~l processing. Typically a sample of tissue 2.C to 2.5 square centimeters in area and 3 to 4 6 ~illimeters thick is utilized. The tissue sample is then ~ixed 7 in a material ta fixative~ which not only preserves the cellular 8 structure but also stops any further enzymic action which couid 9 result in the putrification or autolysis of the tissue. While many substances can function as a fixative, a 4~ formaldehyde or 11 a 10~ formalin solution is very common. Other common fixatives 12 would include ethanol, picric acid or mercuric chloride usually 13 with formalin. It should be ~ red that in dealing with 14 these substances the containers holding the materials must be suitable. For exa~ple mercuric chloride severely ~UL ~ ude~ metals 16 and therefore should normally be contained in a glass vessel.

18 To prepare good samples for microscopic examination the l9 initial step should Xill the enzymic processes of the tissue and should alter or denature the proteins of the cell through 21 fixation. The period of fixation may take several hours or even 22 a few days dPpon~i~q upon the ti6sue type, sample size and type 23 of fixative being used.

After fixation, the tissue sample is often dehydrated 26 by the removal of water from the sample through the use of 27 increasing ~L~n~LI-s of alcohol or of some other dehydrating wog~l33~0 2 1 ~ ~ 7 7 6 r~ Oll~6 l fluid. Gradual dehydration is preferred because it causes less 2 distortion to the sample than a rapid dehydration process.

4 The alcohol is then replaced by a chemical which mixes with wax or some other plastic substance which can permeate the 6 tissue sample and give it a consistency suitable for the 7 preparation of thin sections without disintegration or splitting.
8 Fat solvents, such as chloroform or toluene are commonly used for g this step. The sample, which has been dehydrated by the infiltration of alcohol, is next exposed to several changes of ll solvent over a period that may last from a few hours to days 12 until the alcohol is completely replaced by the solvent. The 13 sample is then exposed to a wax which is soluble in the solvent.
14 lf a paraffin type wax is used the infiltration is at a t~ aLu~~ above its melting point. After the wax infiltration 16 the sample is allowed to cool and the wax solidify so that the 17 sample is entirely ~ in and infiltrated by the wax.

19 A microtome is then ~,til;z_d to cut thin slices from the tissue sample. The slices are on the order of 5 to 6 microns 21 thick. The cut thin sections are floated on water to spread or 22 flatten the section. The section is then ~i~p-~s-d on a glass 23 slide, usually measuring about 8 by 2.5 millimeters.

The wax is then removed by exposing the sample to a 26 solvent, the solvent removed by alcohol, and the alcohol removed 27 by decreasing the alcoholic ~u..~ ations until eventually the 28 tissue is once more infiltrated by water. The infiltration of W09s/332~) 2191776 r ~ .c6 1 the sample by water permits the staining of the cell constituents 2 by water soluble dyes.

4 Prior to the devol~, ~ of automated ~Lo~duLes for the preparation of tissue samples, it often took from 2 to 10 6 days before the tissue could be Py~m i nod under a mi~Losc~ye. In 7 more recent years automated procssses have been developed 8 utilizing apparatus to transfer the sample from one fluid to 9 another at defined intervals, and as a result the preparation time has been significantly reduced to between about 4 and 16 11 hours.

13 Variations in the materials used in the preparation of 14 the sample are advantageous under some circumstances. The use of ester wax allows sections 1 to 3 microns thick to be cut with 16 less contraction than that which occurs when paraffin used. The 17 sample is exposed to higher temperatures when paraffin wax is 18 used. The use of cellulose nitrate omhea~ i ng shrinks tissues 19 less than wax, ~LOdU~eS good cohesion between tissue layers and permits large undistorted sections to be cut 25 to 30 microns 21 thick, if so desired. It is clear that persons with skill in the 22 art of ti8sue pl~a~tion may use many different materials to 23 which the samples may be exposed.

Tissue staining is a pLuceduL~ which is utilized to 26 make microscopic structures more visible. Perhaps the most 27 common stain materials are hematoxylin and eosin. Hematoxylin is 28 utilized to clearly stain the nuclei of cells dark blue. Eosin WO 9513324(1 2 ~ 9 1 7 7 6 r IIL .,, . 15() 1 is used to stains the cell cytoplasm various shades of red or 2 yellow, presenting a clear contrast to the blue stain of the 3 nuclei.
S Many synthetic dyes are derived from benzene which is - 6 colorless but by changing its rh~micAl configuration color 7 , _ '- are ~uduced which are called chl ,'nres It is 8 these ~ res which constitute the bulk of the different 9 coloring dyes used in research and routine histology.
11 There are many techniques by which sample tissues may 12 be stained and most of these techniques require exposing the 13 sample to various solutions. HistArh~-;ctry is the science by 14 which rh~_;eAl reactions are used to identify particular substances in tissues. In addition, many enzymes can be detected 16 by ~Yros; ng a sample to a particular rh~-i ral substance on which 17 the enzyme is known to have an effect such as turning the 18 substance into a colored marker. Thus from the above it can be 19 seen that a sample tissue may be exposed to various antibodies, enzyme labeled detection systems, colormetric substrates, 21 counterstains, washing buffers and organic reagents.

23 Many experimental and obseLv~Lional research projects 24 involve experimentation to authenticate new techniques and these experiments can be very extensive and time concll~ing.

27 In addition to the techniques that prepare samples for 28 optical miuLuScu~y~ techniques often must be utilized which make WO9/33240 2 I q 1 7 7 6 A ~ I/U~ fi 1 the use of electron microscopes suitable in the examination of 2 tissue samples. Actually it has been found that the pathological 3 examination of almost any disorder nakes electron mi~.os~
4 highly desirable and often essential.

6 Tissue samples for use with an electron microscope may 7 be fixed in glutaraldehyde or osmium tetroxide rather than in the 8 standard fixatives used for optical microscopy samples. Usually g very small samples of tissue are ~ d~ in methacrylate or epoxy resin and thin sections are cut (about .06 microns thick).
ll Staining is most often done by colored solutions and not dyes, 12 and heavy metal salts are utilized to enhance contrasts of 13 density.

From the above brief description of some of the 16 techniques and materials used by a pathologist in the examination 17 of tissues, it can be seen that for a research laboratory to 18 carry out such a wide variety of p~cesses and numerous different l9 tests assisting apparatus would be desirable and almost mandatory. Other and further information about tissue analysis 21 and tissue assays may be found in the following references, each 22 of which is hereby inc~L~L~Led by reference as if fully set 23 forth herein:

Bancroft, J.D. and A. Stevens. Theory and Practice of 26 Histological Techniques t3rd ed. 1990). Churchill 27 Livingstone: Edinburgh. ISBN 0-443-03555-8.

21 ql 776 W09sl33240 ~ ' 156 .

1 Childs, G.W. Immunocytnrh~ic~l Technology (1586). Alan R.
2 Liss, Inc.: New York. ISBN 0-8451-4Z13-5.

4 Culling, C.F.A., R.T. Allison and W.T. Barr. Cellular Pathology Technique (4th ed. 1985). Butterworths: London.
6 ISBN 0-407-72903-8.

8 Sternberger, L.A. r - y~n~h~mictry (2nd ed. 1979). John 9 Wiley & Sons: New York. ISBN 0-471-03386-3.
11 Many pathology laboratories have in fact automated many 12 of the simple and routine pluceduL~s described above such as 13 simple staining or sample ~h~;ng. Where the same proceduLe is 14 repeated with great frequency, laboratories have often d~ciqn~d specialized machines to perform the often repeated testing 16 simultaneously on many samples. Typical of such machines are the 17 equipment used in the routine analysis of blood samples. The 18 equipment used in this type of laboratory is capable of treating 19 multiple samples simulr~neo~lc1y to the same testing ~LoceduLe, i.e., parallel testing or through the use of multiple ~-~h;n~c 21 the same result of parallel testing, is achieved. Alternatively 22 the laboratory may perform the same test repetitively, i.e., 23 sequentially and thus subsequent samples may be subject to a 24 significant time delay.
~ 26 Research laboratories often are required to perform 27 non-routine analysis requiring many different test pLOCedUL~S.
28 As a result of this lack of repetitive yLvceduL~ research W09~l33~0 2 ~ 5 i 77 6 r~ c~

1 laboratories have relatively little automated equip~ent to assist 2 the researchers in their task. The most obvious reason for this 3 lack of automation is that the equipment presently available is 4 dedicated to a limited number of ~Loce~uLes most commonly performed. The equipment i6 not flexible enough to permit a wide 6 variety of operations to be easily accomplished nor does the 7 present equipment permit easy and facile changes to the 8 operations.

Another problem that has arisen in the art of repeated 11 testing is that of reagent supply. Typically, devices to perform 12 repeated testing must be loaded with bulk reaqents, and those 13 bulk reagents must have su~ficient volume that a speci- - slide 14 can be i --~sed in the reagent, at least to the level of the speci . This can be wasteful of expensive reagents. It can 16 also result in substantial contamination with the reagent of the 17 back or sides of the slide, resulting in significant ~LLy~vLL of la the reagent and its Ch~iC~l effect into a next step, and a 19 possible safety hazard for the operator or support personnel.
21 Another problem that has arisen in the art of repeated 22 testing is that of packaging of reagents for tests. Typically, 23 devices to perform repeated testing comprise isolator pads, 24 essentially hydrophobic surfaces of glass or plastic, with rol~gh~n~d areas to contain the reagent and smooth areas to repel 26 it. This can cause two problems. First, if too much of the 27 reagent is doled out by the up_L~Lu~, it can overflow the 28 isolator pad and mix with another reagent. Second, the reagent WO 9S133240 ~ 2~.~ q~l ~ 7 6 . ~u~ .'(''1~6 1 has a near maximal surface/volume ratio, often resulting in 2 significant evaporation of the reagent before use.

4 SUM~ARY OF THE INVENTION

6 The invention provides a system which performs a 7 plurality of ;n~r~n~nt analysis procedures simultaneously, 8 possibly involving differing types of tissues and differing 9 process steps. The system comprises a robotic arm, which may move the different tissue samples among a plurality of processing 11 stations, and a processor, which may select the next tissue 12 sample to move, when to move it, and where to move it to. In a 13 preferred ~ nt, the pL~cessor may direct the robotic arm to 14 interleave the differing process steps, for example by time division multiplexing.

17 In a preferred ~ , the proc~ccinq stations may 18 be ~;Cp~s~d in a set of grid locations, so that the location of l9 any one proc~-c;nq station may be specified by an X coordinate and a Y coordinate, and possibly a Z coordinate for height. The Zl robotic device may comprise a bench robot with sufficient degrees 22 of freedom that it is able to reach each of the grid locations 23 with suitable r v~ 8. The processing stations may comprise 24 workstations for performing individual steps of the tissue assay ~l~ceduLes, such as solution trays, or other equipment useful in 26 bioassay, biomedical or related environments.

~'O 95f33240 ~ ~ q l 7 7 ~ Pc IIIJ~,r~C 156 In a preferred e~h~.~i L, the processor may select a 2 tissue sample to be moved in response to timing information about 3 the p~ooedu~~s, which may specify a time duration range (e.g., a 4 minimum time and maximum time) each process step should take.
5 The pLocessor may determine the exact time for a step by 6 qenerating a possible sPqu~n~e of steps and ~-Y~nining that 7 sequence for conflicts, adjusting that sequence in response to 8 those steps with a specified range of times, and iterating the 9 calculation over a plurality of possible sequences. The 10 pLucessoL may also cptimize the order in which samples are moved 11 to minimize the total time required by the system to complete the 12 ~LU'_ dllLt:S, for example by generating a plurality of possible 13 sequences, evaluating each sequence for total expected time, and 14 selecting the best sequence available.

16 In a preferred nmhr~ t, the robotic device comprises 17 a aet of standardized pa~ gPc, ~licro5l d by means of a set of 18 spring locks on a set of standardized tiles and ~ccpc8r~cl by a set l9 of standardized holders for standardized slides or slide pairs, 20 having contents comprising a standardized reagent, chemoactive or 21 bioactive compound or mixture, or buffer, and a set of 22 pLe}LuuL -~ assay protocols. A standardized workstation may 23 also comprise another type of device for operating on sample 24 slides (or other carrying media such as test tubes or wafers~, 25 such as a centrifuge, diffusion, distillation or other separation 26 device, a DNA cro~clinl~ing device, an eleu~LuyvLctor, a microwave 27 device or other radiation source, an incubation oven or other 28 heating unit, or a refrigeration element or other cooling unit.

W~5l33240 ~1 ql 776 .~ 156 1 Because the packages, tiles, contents, and protocols are 2 standardized or preselected, the operator may quickly insert the 3 packages into the tiles, open the packages for operation, and 4 select a pL~LU~L ' assay protocol. All these operations may be performed quickly and may promote rapid and efficient 6 operation of the robotic device.

- In a preferred ~mho~ir L, the processor may comprise a 9 graphic interface by which an operator may specify the steps of a pLùceduLa. A display of the grid locations may comprise symbols 11 for the workstations, which an operator may identify with a 12 pointing device such as a mouse. The operator may create or edit 13 templates for workstations, create or edit lists of process steps 14 for pLuceduLes, monitor the pLoyLess of ongoing procedures, or override the determination of what process steps to perform. For 16 example, in a preferred ~rho~;- L, the operator may create a 17 list of process steps for a procedure by selecting a sequence of 18 workstations with the mouse, and associating timing or other 19 information for each process step with the selected workstation.
The operator may also choose to select a stored list of process 21 steps for a LIL UU~dUL ~ .

23 ' Thus, the invention provides apparatus and methods 24 whereby a plurality of test pLoueduLas can be performed on several samples, e.g., through the use of time division 26 multiplexing. The invention also provides apparatus for use in a 27 laboratory for assisting in the performance of multiple tests 28 which can be easily pLU~L ~ by the operator to execute W09s/33~ 7 7 6 r~ slsfi 1 sequentially timed step procedures for a plurality o~ test 2 samples. The invention also provides a flexible laboratory 3 testing system which may use time division multiplexing to 4 interleave the multiple steps of a plurality of test p~o~du ~s to allow for a plurality of different pLoceduLes to be performed 6 on several different test samples in parallel.

8 BRTFF DESCRIPTION OF T~E DRAWINGS

Figure 1 shows a robotic device for use with the 11 invention.

13 Figure 2 shows a laboratory setup having robotic 14 equipment like that shown in figure 1.

16 Figure 3A shows a standardized tile for coupling to the 17 robotic device.

19 Figure 3B (comprising 6 parts, individually figures 3B-1, 3s-2, 3B-3, 3B-4, 3s-5, and 3B-6) shows a standardized packaqe 21 for coupling to the tile.

23 Figure 3C (comprising 3 parts, individually figures 3C-24 1 and 3C-2, and multiple page figure 3C-3) shows first and second 25 standardized slide holder for coupling slides to a _ ' or 26 mixture in a package.

Wos~/33~u ~ '2 1 9 1 7 76 P../~ 6 1 Figure 3D (comprising 2 parts, individually figures 3D-2 1 and 3D-2) shows a workstation having an incubation oven and a 3 carrying medium for inserting slides or slide pairs.

Figure 3E is a flowchart of a preferred ~ethod of 6 operating the robotic system with standardized packages and 7 contents.

9 Figure 4 is a flowchart showing a time line for five 10 tasks.

12 Figure 5 is a flowchart illustrating multitasking of 13 the tasks shown in Figure 4.

Figure 6 shows a multitask monitoring screen as viewed 16 by an operator.

18 Figure 7 shows a template bnil~i ng screen as viewed by 19 an operator.

21 Figure 8 shows a process bnil~;ng screen as viewed by 22 an operator.

24 Figure 9 shows a process timing screen as viewed by an 25 operator.

W095~33240 21 q ~ 7 7 6 r~ s6 3 Inventions described herein may be made or used in 4 conjunction with inventions described, in whole or in part, in the following patents, publications, or cor~nAinq applications, 6 all of which are hereby incuL,uL~ted by reference as if fully set 7 forth herein.

g U.S. Patent Application Serial No. 07l740,285, filed August 5, 1991, in the name of inventors Steven A. ~ernstein and 11 Page A. Erickson, titled "Method and Apparatus for Automated 12 Tissue Assay"; and 14 U.s. Patent Application Serial No. 08~218,143, filed March 24, 1994, in the name of i,.v~nLoL~ Steven A. Bernstein and 16 Page A. Erickson, titled "Nethod and Apparatus for Automated 17 Tissue Assay".

19 In a preferred P~hoA; L, a multiple axis bench top robot is located to reach peripheral auxiliary equipment ~icpos~d 21 in the operational area of the robot. The robot may respond to 22 the output of a PC type c ~ t ~r which utilizes process control 23 pLO~3 and assay dev~l , L software. Peripheral equipment, a 24 plurality of work modules or workstations, is ~;~pos~ in a grid like pattern around the bench top robot. The workstations may be 26 ~;epncP~ or dLL~ d im any convenient pattern and may be 27 l~pL~~~ ed by a template. Each grid location may contain the woss/3324o '' 2'1 ~ 776 ' P~ 156 1 77ecP~7s;7ry equipment to perform a single step of a tissue assay 2 ~JL VCe~lUL C: .

4 For example, a workstation at a grid position may contain a solution tray into which one or more slides may be 6 i ~ed by the robotic equipment. The slide, or slides, could 7 be i ~ed to a predetermined depth and retained in the solution 8 tray for a precise time. It should be clear that each grid 9 location may have a solution tray having different depths or different ~7.ir ~ir7n~. Alternatively, a grid location could 11 contain a slide holder or other peripheral equipment capable of 12 performing a single function on the sample.

14 The robotic oqlli t or robotic arm may be controlled by a standard PC computer. The assay devel~, t software is 16 graphic in nature and places a model of the peripheral grid on 17 the screen of the computer. While each tissue assay may have all 18 its steps pl~L~yL ' the assay development software permits l9 the steps of the pLVCedULr or the timing of the steps to be altered. The graphic nature of the presentation permits 21 laboratory personnel to alter such elements without the necessity 22 of relying on a computer or ~-oyL~-~ing expert.

24 The process control software associated with the PC may monitor the pLu~LeSs of the assays, may permit manual override of 26 an automatic operation, and most importantly, may permit 27 5~ho~h71 i77g of multiple assays simultaneously in parallel through 28 the use of time interleaving of the various steps in the test W095133240 ~ 9 1 776 r~ . IS6 1 pLOCedl're5. Thus while sample ~1 may be ~icpOc~ at a 2 workstation in a grid location where it undergoes a drying 3 operation, sample #2 may be located in a tray containing a 4 staining solution while sample #3 is undergoing a fixation step.
The timing of each step is accurate and the system interleaves 6 the steps and nt i 1 i 70C the "waiting" or processing time between 7 steps in a single procedure to perform operational steps on other 8 samples which ~ay be undergoing completely different preparation.

LABORATOR~ BENCH AND RO~OTIC 3EVICE

12 Figure 1 shows a robotic device for use with the 13 invention. Figure 2 shows a laboratory setup having robotic 14 P~ nt like that shown in figure 1. The equipment may include a robotic device 10 mounted on a standard laboratory bench top 16 11. The bench top 11 defines the operational area reachable by 17 the robotic device 10. The bench top 11 may have integral 18 therewith a plurality of locating P1- ~ such as holes 12.
19 Alternatively, the locating elements may be ~;~pDsed on a separate base ~;~posPd between the robotic device 10 and the 21 laboratory bench top 11. A template may be used to ~Lese~l~ the 22 operational area and to assist in defining the eXact location of 23 each workstation.

Located on the bench top 11 are one or more work 26 modules 13. A control station 14 is located adjacent to the 27 laboratory bench 11. The control station 14 may include a 28 typical PC type camputer 15, such as an IBM , -tible co~puter wogsl33240 2 1 9 ~ 776 P~ 6 1 having an Intel x86 ~Locessuc, or a computer similar thereto, 2 mounted on a desk 16 or other working surface. It would be clear 3 to one of ordinary skill in the art, after perusal of the 4 specification, drawings and claims herein, that other types of computers may be utilized to control the movement of the robotic 6 arm 10. A printer 17 is shown although other peripheral 7 equipments may be utilized in conjunction with the computer 15.

9 Referring to the bench top 11, a plurality of locating holes 12 are ~i CpOSG~ at predetermined fixed locations relative 11 to the robotic device 10. The locating holes are designed to 12 receive modular workstations 13. Each modular workstation 13 is 13 designed to be used in the performance of a particular process or 14 step in one laboratory task or test ~L O~dUL ~ . Thus each function required to be performed in a task is associated with a 16 work module 13 which has a pr~ p~ known position on the work 17 bench 11.

19 There are a number of methods by which the location of a particular work module 13 can be supplied to the computer 10.
21 For example each work module 13 may include a floppy disk which 22 would contain the physical characteristics of the work module, 23 such as its height, width and length. The customized data for 24 each module would be fed into the central processing unit of the computer and would query the operator, for example through a CRT
26 display, to provide the location of the work module. The 27 operator through the keyboard input would specify the location of 28 the module on the locating grid. Thus for each work module or W095~332~0 ~3i9 1 776 ~ S~
.

1 step of a task the computer would have stored in its memory the 2 physical characteristics and location of the module.

4 In a preferred : ' '; L, the robotic device 10 is S capable of travel in an X direction along a first cable driven 6 bearing 20 (actuated by a first cable drive 20a1. Disposed at 7 right angle to and vertical with respect to the first cable 8 driven bearing 20 is a second cable driven bearing 21 (actuated 9 by a second cable drive 21a), capable of traversing the first cable drive 20a. Coupled to the cable drive 21a is a third cable 11 driven bearing 22 (actuated by a third cable drive 22a~ pos~d 12 at a right angle. A robotic hand 23 is mounted on cable drive 22 13 and comprises a spring loaded solenoid 23a coupled to a rubber 14 securing ring 23b. The securing ring 23b is capable of coupling to a sample carrier 23c. The sample to be assayed (which may be 16 a tissue sample) is mounted on the sample carrier 23c.

18 Thus the hand 23 on which the sample is mounted is 19 capable of X ~. ' along cable driven bearing 20, Y ~,~c t along cable driven bearing 21, and Z ~v~ -t along cable drlven 21 bearing 22. The system illustrated is thus capable of motion 22 relative to three axes. Although the system is illustrated using 23 cable driven bearings 20, 21 and 22, it would be clear to those 24 skilled in the art, after perusal of this application, that other robotic equipment could be provided that could decrease or 26 increase the number of axes, that other techniques other than 27 cable drives and cable driven bearings, ~such as lead screws, 28 gears, belts, or other devices) could be used, that such other W09sl332~0 ~2 1 ~1 7 76 ~ 56 ~ nt or techniques would be workable, and are within the 2 scope and spirit of the invention.

4 Typically, the range of movement along the X axis may be about 76 inches, along the Y axis about 19 inches, and along 6 the Z axis about 18 inches. Such a typical range of v, --t 7 could provide approximately 15 cubic feet of operational area.

g SYSTEM OPERATION

11 In order to illustrate the operation of this invention, 12 let it be assumed that the laboratory has five example tasks to 13 accomplish, each having five example steps. For purposes of 14 illustration, the five steps in each of the five tasks will be utilized to d LL~te the multitasking capabilities of the 16 invention. The five tasks and the five steps of each of the 17 tasks are shown in Table 1 herein.

19 It is apparent from Table 1 that some of the tasks utilize the same steps such as Pad 1 or suffer 1. If these steps 21 were to be carried out in accordance with the principles of this 22 invention, it would be n~c~cs~ry to provide only 14 work modules 23 even though 25 steps were being perfor~ed. Disposed on the grid 24 would be a separate work module for each of the 14 different steps listed above. Thus there would be a Pad 1 module to be 26 used in carrying out seven of the above steps. Alternatively, 27 the user could provide multiple modules, each capable of 28 performing the pad function. A Buffer 1 module would be used for W095~3240 2 1 9 1 7 7 6 r. ~ l56 Task #1 Basic Fuchsin staining 2 Step #1 Buffer 1 Step #2 8uffer 2 3 Step ~3 Basic Fuchsin Step ~4 Pad l 4 Step ~5 Buffer 2 Task #2 Azure II & Methylene Blue Counterstaining 6 Step #1 Azure II
Step #2 Pad 1 7 Step #3 Buffer 1 Step #4 Pad 1 8 Step #5 Nethylene Blue 9 Task ~3 Tissue Fixation Step #1 Isotonic Rinse Step #2 Primary Fixative Step #3 Buffer 1 11 Step ~4 Buffer 2 12 Step ~5 Secondary Fixative Task #4 Immunocytochemistry 13 Step #1 Buffer 1 Step #2 Pad 1 14 Step #3 Blocking Antibody Step f4 Pad 1 Step ~g Buffer 1 16 Task #5 Slide Sil;ni~;ng Step #1 APTES
17 Step f2 Toluene Step ~3 Water 18 Step ~4 Pad 1 l9 Step ~5 Oven five of the steps and a Buffer 2 module for two of the steps.

Each of the rr-~inin~ steps would have a module ~;Cpoc~d on the grid to perform tho n~ ry work associated with the step.

It is often essential that the step of the task be performed within certain time limits. The timing of some steps can be critical. Figure 4 is a flowchart showing a time line for the five steps of the tasks in Table 1. It should be noted that Task ~l, Step #1 ~ -nr~c at g:00 and has a duration of ~ W095~33240 21~91 7 76 r~ s6 1 approximately 15 minutes, inclusive of the time n~ri c~ry to 2 transport the sample to the location where Step #2 is performed.
3 Thus Step ~2 will re at approximately 9:15. It should be 4 noted that the timing for the start of Step #2 has some leeway in that it can ~ -e between 9:15 and 9:18, providing leeway of ~ 6 three minutes. Step #2 has a duration of approximately 11 7 minutes and the sample is transported to the location where Step 8 #3 will be performed. The time for performing Step #3 is 9 critical as indicated by the lack of interval for the starting times. Step #3 must _ -nre at 9:26. Fourteen minutes later 11 the sample is undergoing Step #4, which can -- any time 12 between 9:40 and 9:50. The last Step #5 is performed at 9:51.
13 It should be noted that if each Step is -nrP~ at the outer 14 time limit Step #5 may not begin until 10:22.

16 In a similar manner it can be determined from figure 4 17 that the five steps of Task #2 may consume 1 hour 34 minutes, 18 Task #3, 1 hour 9 minutes, Task #4, 1 hour 17 minutes, and Task 19 #5, 1 hour 16 minutes. Thus if the five steps of the tasks shown were to be performed se~uentially the total time to completion 21 would be 6 hours 38 minutes.

23 Referring to figure 5, the multitasking method of this 24 invention is therein illustrated to show the time interleaving of the steps of the multiple tasks. Acsnm;ng again for purposes of 26 illustration and simplification of explanation that we are 27 desirous of performing the same five steps for the same five 28 tasks. Under the control of the computer the robotic hand would W0~332~0 ~ 2l ql 7 7 6 ~ 6 1 be _ n~ to obtain sample #1 or alternatively the sample 2 could be brought to the robotic hand and for grasping. The hand 3 retaining the grasped sample would move the sample to the 4 location of the work module for Task ~1, Step #1, i.e., Buffer 1.
The sample would be freed from the hand and left at the work 6 module. The hand would proceed to the location of sample #2 7 where it would grasp the sample and carry it to the work station 8 where Task t2, Step #1 would be performed.

Each of the five samples would in turn be grasped by 11 the robotic hand and transported to the work module associated 12 with the first step of the task to be performed on each sample.
13 It should be noted that the design of the Buffer and Pad work 14 modules permit the simultaneous treatment of at least two samples from different tasks. Alternatively, two work modules could be 16 provided so that each sample could be treated in a different 17 module.

After locating sample #5 in the Task ~5, Step tl module, the robotic hand returns to the module for Task X5, Step 21 #1 and gasps the sample #5 and transports it to the module for 22 Task t5, Step #2. Following the path ill~L~ted in ~igure 5, 23 the hand proceeds from the Task #5, Step ~Z module to Task #3, 24 Step #3 module where it grasps sample #3 and transports it to Task #3, Step #2 module where the sample is deposited. The hand 26 then returns to the location of the first sample which is in the 27 module associated with Task tl, Step tl and takes it to the 28 module for Task #1, Step #2. The hand returns to the location ~ W09s~33~ 2 1 9 1 7 7 6 P.~ 56 1 sample #4 and carries it to Task #4, Step #2 and then at the 2 appropriate time transports the same sample to Step #3 of Task 3 ~4.
At this point in the operation of the system, the 6 computer detects that Task #1, Step ~3 and Task #2, Step #2 are 7 both scheduled to start at the same time, 9:26. In order to 8 resolve the conflict the system utilizes a technique, herein 9 termed "fuzzy timing", to process the control of the robotic hand and optimize the process. Fuzzy timing may comprise the window 11 of time during which each process (Task) step may occur without 12 affecting the process results. Some steps of a process may be 13 critically timed, i.e., the time required for that step is exact, 14 such as Task #1, Step ~3 in figure 5, but in general most steps a process the timing is less critical and may comprise any amount 16 of time within a known range and thus are noncritical in their 17 timing, such as Task #2, Step ~2, which has a window of four 18 minutes, as shown in figure 5. The system of this invention uses 19 these windows of time to advantage as to optimize ~minimize) the time n~c~Cc~ry to complete the multiple tasks.

22 The use and advantages of "fuzzy timing" can be 23 illustrated by considering two different tasks, each having a 24 process step terminating at the same time or within moments of the another. Acs~ g that both steps are critically timed in so 26 far as the termination time is c~ eL~Ied~ it is apparent that 27 both samples from the two different steps can not be moved to the 28 next step in each process simultaneously since cu,l~uLL_.lL

W095l3324~) ~~2~1 ~ 1 7 7 ~ .~"~

~ L of two samples is not within the capabilities of this 2 ~mho~ '. Thus it is necessary to adjust the starting times 3 for the two steps relative to each other so that the ending times 4 will allow for the - ~ -n~ of each sample to its next process step. While this can be done quite easily, it is clear that the 6 mere adjustment of a starting time for a step in the process may 7 well cause other timing conflicts. It is possible that under 8 such conditions the system could not support simultaneous 9 throughput of multiple ~rucecses unless the timing was altered.

11 Fuzzy timing allows the system additional flexibility 12 since by providing a window of time at each noncritically timed 13 process step, conflicts will be minimi7Qd through the adjustment 14 of timing at the step level, rather than by shi~ting the timing of the whole process or task.

17 STA~DARDIZED CHEMICALS AND PACKAGES

19 Figure 3A shows a standardized tile ~or coupling to the robotic device.

22 As described herein, the robotic device 10 may be 23 mounted on a bench top 11 having a plurality of locating elements 24 such as holes 12 and having a plurality of work modules 13 ~i Cp~CQ~ thereon.

woss/332~0 ~ 7 7 6 . ~ r~ls6 1 In a preferred Pmho~ir t, each work module 13 2 coDprises one or more tiles 301, each tile 301 comprising a 3 molded plastic piece having a top face 302 and a bottom face 303.

S The bottom face 303 of the tile 301 comprises a 6 relatively flat plastic surface 304, possibly having one or more 7 bottom indentations 305 and bottom ribs 306, and having a set of 8 receiving wells 307 for insertion of a CG~r l~on~ing set of 9 fasteners 308. As shown in figure 3A, the fasteners 308 fit through a set of holes 12 for a designated location on the bench 11 top 11, and are coupled to the receiving wells 307 for fastening 12 the tile 301 to the top surface of the bench top 11.

14 In a preferred ~ho~i t, the fasteners 308 comprise screws, but those skilled in the art will recognize, after 16 perusal of this application, that other types of fasteners would 17 also be workable with the devices and substances described 18 herein, and are within the scope and spirit of the invention.

The top face 302 of the tile 301 comprises a set of 21 receiving areas 309 for insertion of a ~uLL~onding set of 22 standardized p ir~g~e 401. The top face 302 also comprises a set 23 of one or more top indentations 310 and top ribs 311. A set of 24 holes 312 are ~iCp~CP~ in at least some of the top indentations 310, so that liquids in those top indentations 310 may drain.
26 Each receiving area 309 comprises a depression 320, into which a 27 package 401 (figure 3B) may be placed.

WO~.S/33241~ 2 1 ~ 1 77~ 6 1 Each depression 320 comprises a pair of side walls 321 2 d;cpos~d parallel to each other, a pair of in~ermediate barriers 3 322 di Rpocqd 50 as to dLvide the depression 320 into a set of 4 three subdepressions 323, each intermediate barrier 322 having a S pair of stubs 324. Each pair of stubs 324 is aligned with each 6 other and ~icpnc~d parallel to the side walls 321, so that a 7 package 401 may be snugly fitted into one of the three 8 subdepressions 323.

Each stub 324 comprises a first and second stub side 11 325 and a stub end 326. The stub sides 325 for the stub 324 are 12 ~;CrO5oA parallel to the stub sides 325 of the matching stub 324, 13 and parallel to the side walls 321. The stub end 326 for the 14 stub 324 is generally d;cposod so that the stub 324 is relatively short compared with the package 401.

17 ~hen a package 401 is fitted into a side one of the 18 three subdepressions 323, it is ~tsp~cPd with a first package 19 side 402 ~figure 3B~ d;cp~cod next to a first side wall 321 and with a second package side 402 d;cp~5~ next to one of the stubs 21 323, in particular, next to one of the stub sides 324. A first 22 end of the second package side 402 is d;~posod next to a first 23 stub 323, while a second end of the second package side 402 is 24 d;cp~s~d next to a second stub 323, the second stub 323 being the matching stub 323 aligned with the first stub 323.
26 ..
27 Alternatively, a package 401 may be fitted into a 28 center one of the three su~depressions 324. In this case, it is Wossl332~o ~ 2 1 q 1 7 76 r~ G

1 ~iqposPd with a first package side 402 disposed next to a first 2 pair of stubs 323, and a second package side 402 next to a second 3 pair of stubs 323. A first end 403 (figure 3B) of the second 4 package side 402 is ~;qp~s~d next to a first stub 323 in its pair, while a second end 403 of the second package side 402 is 6 ~i RpO5P~ next to a second stub 323 in its pair, the second stub 7 323 being the matching stub 323 aligned with the first stub 323.

9 Each subdepression 323 comprises a pair of receiving holes 325 for insertion of a cUL~ nsing lever 404 (figure 3B) 11 and a cuLIo-~u~ tng spring lock 405 (figure 3B) of the package 12 401 to be ~icpnsod in the subdepression 323. When the package 13 401 is fitted into the subdepression 323, the lever 404 of the 14 package 401 is ~icposed in a first one of the receiving holes 325, and the spring lock 405 of the package 401 is d;cp~sPd in 16 the second one of the receiving holes 325.

18 Figure 3B (comprising 6 parts, individually figures 3B-19 1, 3B-2, 3B-3, 3B-4, 3B-5, and 3B-6) shows a standardized package for coupling to the tile.

22 In a preferred ~ L, a standardized package 401 23 comprises a molded plastic tray 406 and a thin cover 407 affixed 24 to the tray 406, such as by a heat weld, a glue, or other known means. In a preferred P~ho~ t, the thin cover 407 may 26 comprise a plastic or metallic sheet 408, laminated on an outside 27 side 409 with plastic and printed thereon with identifying 28 information, and coated along an edge area 410 on an inside side WO~S/3324~ 2 i 9 l 7 7 6 r~ s6 1 411 with a fixative 412 and affixed by means of that edge area 2 410 to a CULL~ r n~ir~ tray surface 413.

4 In a preferred ~ho~;~ L, the fixative 412 comprises a heat weld, but those skilled in the art will recognize, after 6 perusal of this application, that other types o~ bonding 7 techniques would also be workable, such as crimping or welding, 8 or glue, and are within the scope and spirit of the invention.

The tray 406 comprises a tray frame 420, having a 11 rectilinear shape with a top surface 421. The top surface 12 includes the tray surface 413 for bonding with the cover 407, and 13 also in~ln~pc a handle region 422 with a hole 423 ~icrrsed 14 therein.

16 The cover 407 also comprlses a cover ~ip 414 r~li cpo6~
17 on at least one end cf the package 401, having a sufficient size 18 to be grasped by an operator and re~oved from the tray 406.

The tray frame 420 comprises a pair cf side surfaces 21 424, ~iCpoCP~ perpendicular to the top surface 421. The side 22 surfaces 424 form the package sides 402 and the ends 403 of the 23 packet sides 402.

The tray frame 420 comprises a first end surface 425, 26 r1;Cpospd perp~n~ir~ r to the top surface 421 and to the side 27 surfaces 424, and for~ing a bo~ shape underneath the handle wog5~3324u ~ ~ ' ~ 1 7 76 ~ 6 1 region 422 and the hole 423, providing additional sturdiness in 2 that region.

4 The tray frame 420 comprises a second end surface 426, ~; CpOSD~ perpendicular to the top surface 421 and to the side 6 surfaces 424, and having the spring lock 405 ~icpOS~ thereon.

8 The tray frame 420 comprises a set of tray ribs 427, g ~;cposed underneath the top surface 421 and near the side surfaces 424, providing additional sturdiness to the tray 406 and ll the side surfaces 424.

13 The tray frame 420 is coupled to a well frame 440, 14 which comprises a rectilinear shape having a pair of well sides 441, a well bottom 442, a set of wells 460, a first well end 443 16 near the first end sur$ace 425, and a second well end 444 near 17 the second end surface 426.

19 The wells 460 each comprises a truncated wedge shape, having a single well bottom 461 that is U-shaped, with the plane 21 of the U-shape parallel to the side surfaces 424, and a pair of 22 single well sides 462 that are flat and each have a trapezoidal 23 shape. Each single well bottom 461 comprises a set of three 24 relatively ctraight surfaces, a well horizontal bottom 463 that is relatively flat and horizontal (and may comprise a V shape 26 with a arms of the V shape ~;ep~e~d about 2.5 degrees from 27 horizontal), and a pair of well semibottoms 464 that are flat and 28 ~;qpos~ at an angle of about 9.5 degrees $rom the vertical.

~09s~3240 2 ~ 7 6 F~m~ 156 1 The single well bottoms 461 are ~i$pr~ced in a 2 continuous sequence sb as to merge to form the well bottom 442.
3 The well bottom 442 is therefore formed without seams and with 4 ridges 465 formed by well semlbottoms 464 adjacent to each other.

6 The single well sides 462 are ~i cpo5r~d in a continuous 7 sequence so as to merge to form the well sides 441. The well 8 sides are therefore formed without seams and without ridges.

Each well 460 is formed with a molded label 466 that is 11 unique within the package 410. In a preferred o~ho~; -L, the 12 labels 466 are formed by molding the plastic of the tray 406, but 13 those skilled in the art will recognize, after perusal of this 14 application, that tha labels could be workably formed by alternative means, such as etching, printing, or scoring, and 16 that such alternative means are within the scope and spirit of 17 the invention. In a preferred : i L, the labels 466 may 18 each comprise a single digit uO.., "1", "2", "3", ~4", ~-5U, "6", 19 U7--, "8", or "9". Alternatively, the number nlC" may be substituted for the digit "0".

22 A first well 460 with a label 466 of "0" is ~icposed 23 near the first end sur~ace 425 and has the lever 404 ~;cpo5ed 24 thereon.

26 A second well 460 with a label 466 of n1n is ~iCp~f~
27 near the second end surface 426 and has a set of end ribs 467 28 d; crOSPd thereon.

~ WO9S/33240 ~ 2 1~ q 1 1 7 6 r~ 156 1 The lever 404 comprises a right-angled lever lip 480, 2 having a first lever surface 481 and a second lever surface 482, 3 supported by a set of lever ribs 483 disposed between the first 4 lever surface 481 and the first well 460 and underneath the S second lever surface 482. The lever lip 480 is disposed at 6 parallel to the first end surface 425 and sized to fit into the 7 COLL~ ng receiving hole 325. In a preferred embodiment, the 8 first lever surface 481 has at least one lip hole 484 ~; crosed 9 thereon, to promote mating at a surface of the tile 301 near the receiving hole 325.

12 The spring lock 405 comprises a right-angled spring lip 13 500, having a first spring surface 501 and a second spring 14 surface 502, supported by a set of first spring ribs 503 underneath the second spring surface 502. The first spring 16 surface 501 comprises a section of the second end surface 426 17 having a pair of cuts 504 ~;cp~c~ thereon, a reinforced spring 18 base 505 ~; cpo5~ at a base of the pair of cuts 504, and a pair 19 of second spring ribs 506 ~;qpnsed underneath the second spring surface 426 near the cuts 504. The spring lip 500 is ~;cpOc~ in 21 parallel to the second end surface 426 and sized to fit into the 22 COLL. ~.",.l;ng receiving hole 325.

24 An inside surface 520 of the tray 406 comprises a set of inside wells 521 C~LL~lJ~n~;ng to the wells 460. Each 26 adjacent pair of inside wells 521 is separated by a well divider 27 522. Each well divider 522 comprises a U-shaped, with the plane 28 of the U-shape perpendicular to the side surfaces 424, and having W09sl33240 '~ 21 9 1 776 1 a taper from thicker near a bottom end 523 dicposed near the 2 bottom 524 of the inside wells 521 to thinner near a top end 525 3 ~iFposo~ farther from the bottom 524 of the inside wells 52i.

Each well divider 522 comprises a center 526, at a 6 bottom curve of the U-shape, that has an indentation 527, thus 7 forming two lips 528 di~pose~ between each adjacent pair of 8 inside wells 521.

Each well divider 522 comprises a well top 527, at a 11 pair of top ends 528 of the U-shape, ~;cpn~ed with a gap 529 12 between the well top 527 and the cover 407.

14 In a preferred orhQ~ir-~t, the well dividers 522 are sized so that each inside well 521 may hold 750 microliters (3/4 16 of a milliliter3 of liquid without spilling over to an adjacent 17 inside well 521. However, if the amount of liquid in an inside 18 well 521 exceeds 750 microliters, the liquid will spill over the 19 bottom curve of the U-shape of the well divider 522, and thus spill into the adjacent inside well 521.

22 In a preferred o~ho~i~ L, the robotic device 10 23 ope~ates by orienting a slide 540 with a specimen 541 vertically 24 for insertion into the inside well 521, i.e., with the flat surfaces of the slide 540 being perpendicular to a plane of the 26 ground. When the slide 540 is inserted into the inside well 521, 27 a liquid content 542 of tAe inside well 521 will coat the 28 spoci- - 541 by means of capillary action.

~ Woss/33240 ~ 7 7 6 ' r~ s~

1 This capillary action is particularly promoted if the 2 slide 540 is coupled to a second slide 540 to form a slide pair 3 543, with the spec; 542 sandwiched between the slide 540 and 4 the second slide 540 of the slide pair 543, and with the slide 540 and the second slide 540 maintained a selected separation - 6 distance apart of preferably about 146 microns +/- 12 microns.
7 However, those skilled in the art will r~coqn;ze, after perusal 8 of this application, that slides of differing sizes and selected 9 separation distances would be workable, and are within the scope and spirit of the invention. For example, a selected separation 11 distance for a slide 540 or a slide pair 543 for frozen tissue 12 may comprise a substantially larger size, such as about 200 13 microns. A preferred ~mho~ t of the slide pair 543 is shown 14 in one or more of the following U.S. Patents, hereby inooL~L~ted by re$erence as if fully set forth herein: 4,731,335; 4,777,020;
16 4,798,706; 4,801,431; 4,975,250; 5,002,736; 5,023,187; and 17 5,116,727, and may be used in conjunction with inventions 18 therein.

It has been found by the inventors that the selection 21 of the particular volume, 750 microliters, for each inside well 22 521 is particularly advantageous. This selected volume of liquid 23 is generally sufficient to perform all the steps of typical 24 ; -~ictochemical stains and other assay protocols (generally, with this selected volume of liguid, slides 540 or slide pairs 26 543 may be inserted into the inside well up to about three 27 times). ~owever, this selected volume of liquid is not so large 28 that nonspecimen parts of slides 540 or slide pairs 543 (such as WO9~l332~0 ~ 7 7 ~ r -~56 1 the back or sides~ are regularly excessively c~nt~in~ted. This 2 s~ ted volume of liquid also has the advantage, particularly 3 when held in an inside well 521 having a single well bottom 461 4 with relatively steep sides (formed by the well horizontal bottom 463 and the well semibottoms 464~, that there is a reduced 6 surface/volume ratio. This provides for lesser evaporation of 7 the liquid in the inside well 521.

9 It has also been found by the inventors that the selected shape of the inside well 521 is particularly 11 advantageous. This particular shape promotes self-levelling and 12 reduced ~v~pGr~tiOn, as noted herein. Noreover, this particular 13 shape promotes centering within the inside well 521 of small 14 amounts of liquid tabout 150 microliters), due to surface tension r~p~ n of the liquid by the well semibottoms 464. Centering 16 of the liquid ~LI ~ capillary action when a slide 540 or slide 1~ pair 543 is inserted into the inside well 521.

19 Preferred filling amounts for content of the inside well 521 are about 350 microliters when the _ ' or mixture 21 is not too expensive, and about 200 microliters when the __I-d 22 or mixture is relatively expensive (or when other reasons exist 23 to restrict the amount, such as the e _ ' or mixture being 24 dangerous in quantity).

26 Preferred dimensions and tolerances for tiles 301 and 27 packages 401 are shown in figures 3A and 3B.

W~9s/332~0 ~ie ~ ï-7 7 6 P~ s6 1 Figure 3C (comprising 2 parts, individually figures 3C-2 1 and 3C-2, and multiple page figure 3C-3) shows first and second 3 standardized slide carriers for coupling slides to a , ' or 4 mixture in a package.

6 In a preferred ~mho~;~ L, a first standardized slide 7 carrier 560 comprises a frame 561, a coupling ring 562, a set of 8 slide frames 563, and a set of feet 564. The frame 561 comprises 9 a metal frame comprising a set of four horizontal elements (a top 565, a slide top 566, a slide bottom 567, and a bottom 568), and 11 a set of four support posts 569. The top 565, slide top 566, 12 slide bottom 567, and bottom 568 are coupled and supported by the 13 four support posts 569, to make the frame 561 rigid and sturdy.

The coupling ring 562 is coupled to the top 565 by 16 means of a pair of ring ~U,U,UUL L~ 570, that connect the coupling 17 ring 562 to the rest of the top 565. The coupling ring 562 is a 18 roughly circular element and has a similarly shaped ring base 571 19 underlying it and coupled to it by means of screws 572 ~icposed through the ring supports 570 with their axes aligned vertically.
21 The coupling ring 562 also has a ring bumper 573 ~;cposPd on top 22 and coupled to it by means of glue or another fastening 23 technique.

The coupling ring 562 comprises a plastic disk 574, 26 defining a circular hole 575 (smaller to and aligned with a 27 circular hole 576 defined by each of the coupling ring 562, the 28 ring base 571, and the ring bumper 573), and having a circular 21 ql 776 W0~33240 rcl~u~ 15 1 raised lip 577 surrounding the hole 576. The disk 574 also 2 comprises a circular flat portion 578 ~ierosD~ between the 3 co~rling ring 562 and the ring base 571, sufficiently large so 4 that the disk 574 cannot fall out from between the two. The coupling ring 562 is thus disposed and shaped so the robct's 6 rubber securing ring 23b may couple thereto and form a firm (but 7 easily ~o~hRhle) coupling.

9 If the robot's rubber securing ring 23b is slightly lû misaligned from the disk 574 in the X or Y direction or both, the ll disk 574 will realign within the rubber securing ring 23b by 12 sliding within the region defined between the coupling ring 562 13 and the ring base 571. The rubber securing ring 23b and the disk 14 574 may thus couple anyway despite slight misalignment in the X
or Y direction or both, up to about 2 mm in a preferred 16 ~ r . Similarly, if the robot's rubber securing ring 23b 17 is slightly misaligned from the disk 574 in the Z direction, an 18 outside part of the rubber securing ring 23b will bump against 19 the ring bumper 573, so the rubber securing ring 23b and the disk 574 may thus couple anyway despite sllght misalignment in the Z
21 direction, up to about 2 mm in a preferred ~ t.

23 The slide frames 563 lpreferably there are three of 24 them) are coupled to the slide top 566, by means of a set of screws 580 ~i~posed with their axes aligned vertically. Each 26 slide frame 563 comprises a set of slide positions 581 for 27 holding standardized slides 540 or slide pairs 543. The slide 28 bottom 567 is ~i~p~5Dd to support the slide frames 563 relatively wossl3324o 21 9 1 7 7 6 PCT~S95/06156 1 tightly. An underside 582 of the slide bottom 567 is labelled 2 with a set of letters 583 "A", "B", and "C", ~iRpo~r~ with one 3 letter near each slide frame 563, and a set of digits 584 "1", 4 "2", "3", "4", "5", "6", "7", "8", "g", and "o", ~icposrd with one digit near each slide position 581 in each slide frame 563.
6 A preferred ~mho~ L for the slide frame 563, and related 7 inventions, are shown in one or more of the following U.S.
8 Patents, hereby incu.~u.~ted by reference as if fully set forth 9 herein: 4,731,335; 4,777,020; 4,798,706 4,801,431; 4,975,250;
5,002,736; 5,023,187; and 5,116,727, and may be used in 11 conjunction with inventions shown therein.

13 The set of feet 564 (preferably there are four of them) 14 are coupled to the frame bottom 568, by means of being lntegratedly formed therewith. The feet 564 each comprise a 16 wedge-shaped element 590, with a relatively thicker top end 591 17 and a relatively thinner bottom end 592, shaped and sized to fit 18 into the top indentations 310 in the top face 302 of the tile 301 19 with a bit of extra space.
21 If, when the robot hand 23 deposits the slide carrier 22 560 onto the tile, the slide carrier's feet 564 are slightly 23 misaligned from the tile's top indentations 310 in the X or Y
24 direction or both, the wedge-shaped element 59o will realign within the top indentations 310 by force of the weight of the 26 slide carrier 560, so the slide carrier's feet 564 and the top 27 indentations 310 may thus couple anyway despite slight 28 m;R~ L in the Z direction, up to about 2 mm in a preferred Wog.~/332~0 ~ 9 1 7 7 6 1 embodiment. Similarly, if the slide carrier's feet 564 are 2 slightly misaligned from the top indentations 310 in the Z
3 direction, the slide carrier 560 will fall into the top 4 indentations 310, 50 the slide carrier's feet 564 and the top indentations 3120 may thus couple anyway despite slight 6 misalignment in the Z direction, up to about 2 mm in a preferred 7 ~ho~i L.

9 In a preferred ~ho~i- L, a second standardized slide carrier 600 also comprises a frame 601, a coupling ring 602, a 11 slide frame 603, and a set of feet 604. The second standardized 12 slide carrier 600 comprises a similar structure to the first 13 standardized slide carrier 560.

The first standardized slide carrier 560 comprises a 16 qenerally cubic shape and is adapted for holding a set of three 17 slide frames 563, each with 10 slide pairs ~i.e., a total of 60 18 slides). However, the second standardized slide carrier 600 19 comprises a rectil in~r shape and is adapted for holding a slide frame 603 with 10 slide pairs (i.e., 20 slides).

22 The first standardized slide carrier 560 comprises a 23 roughly circular coupling ring 562, coupled to the top 565 by 24 ~eans of a pair of ring supports 570, which has a similarly shaped ring base 571 underlying it, and which also has a ring 26 bumper 573 ~;~posqd on top. However, the second ~L~..d~rdized 27 sllde carrier 600 comprises a coupling ring 602 that is 28 integrated into the rest of a top 605 and is thus rectilinear, wos~s/3324o ~ 9 1 7 7 6 . ~ I ~U... 5. ~ ' 156 1 which has a similarly shaped ring base 606 underlying it, and 2 which also has a ring bumper 607 r~;spos~ofl on its top. The ring 3 bumper 607 is roughly circular but shaped to match the shape of 4 the second standardized slide carrier 600.

s 6 The first standardized slide carrier 560 preferably 7 comprises a set of three slide frames 563. However, the second 8 standardized slide carrier 600 preferably comprises a single 9 slide frame 608 having a plurality of slide positions 609. The underside 610 of the slide frame 608 is not labelled; rather, a 11 pair of sides 611 of the slide frame 608 are labelled with a set 12 of integers 612 "1", "2", "3", "4", "5", "6", "7", "8", "9", and 13 "10", tlicpo5~rl with one digit near each slide position 609 in the 14 slide frame 608.
16 The first standardized slide carrier 560 preferably 17 comprises a set of four feet 564. However, the second 18 standardized slide carrier 600 preferably comprises a set of only 19 two feet 613.
21 Multiple page figure 3C-3, comprising 13 pages, shows 22 detailed parts drawings for the first standardized slide carrier 23 560 and the second standardized slide carrier 600.

WORKSTATION DEVICES
~ 26 27 In addition to pArkAg~-c 401, the tile 301 at a 28 workstation 13 may be coupled to another type of device for wog533240 ~ ' ~lq~776 ~ 156 1 operating on samples, whether carried by slides 540, slide pairs 2 543, or another carrying medium such as a beaker, test tube or 3 wafer. In a preferred ~ rL~ the tile 301 at a workstation 4 13 may be coupled to one or more of the following devices:

6 The wuLh~Ldtion 13 may comprise a centrifuge, a 7 di~fusion device, a distillation device, or other separation 8 device.

The workstation 13 may comprise a DNA crosslinking 11 device.

13 The WOLhSLdtiOn 13 may comprise an ele~LL~L~tor.

The w~Lhstation 13 may comprise a laser device or other 16 optical device.

18 The workstation 13 may comprise a microwave device, a 19 shielded radioactive sample, or other radiation source, such as a source of ele~LL ~n~tic or ionic radiation.

22 The ~hsLdtion 13 may comprise an incubation oven or 23 other heating unit.

The workstation 13 may comprise a refrigeration element 26 or other cooling unit.

w095/332~0 2 1 9 1 7 7 6 ~ s6 1 Figure 3D (comprising 2 parts, individually figures 3D-2 1 and 3D-2) shows a workstation having an incubation oven and a 3 carrying medium for inserting slides 540 or slide pairs 543.

In a preferred embodiment, an incubation oven 620 6 comprises a chassis 621, an incubation chamber 622 a set of heat 7 oYrh~ngor fins 623, a hydration fluid supply 624, an internal 8 cooling element 625, a fill/drain control 626, a fluid waste 9 receiver 627, a receiving element 628 for a carrying medium 630, and a set of heat fins 629.

12 The incubation chamber 622 is supported by the chassis 13 621 and comprises a set of chamber walls 631 dicrosed in a 14 generally rectilinp~r form 632 with a set of rounded corners 633 to form a first part of a sealed fluid-tight box 634 when the 16 carrying medium 630 is d;cpo5Pd for operation. When the carrying 17 medium 630 is dicprcod for operation, the slides 540 or slide 18 pairs 543 in the carrying medium 630 may be heated with moist 19 heat formed by heating the incubation chamber 622 while disposing a hydrating fluid therein, and thus incubated. Incubation of 21 slides 540 or slide pairs 543 is known in the art.

23 The heat exchanger fins 623 are disposed in the 24 incubation chamber 622 in an array. The array is diqposPd to match, but not contact, a set of slides 540 or slide pairs 543 26 ~iqpospd in the carrying medium 630. There should be one of the 27 heat PYrh~nqor fins 623 for each slide 540 or slide pair 543, or 28 at the least, for each pair of slides 540 or slide pairs 543.

W~9.~l33240 2-! 9 l 7 7 6 ,~ r~

1 Each onc of the heat DY~h~gDr fins 623 has a height sufficient 2 to heat the entire slide 540 or slide pair 543, or at least a 3 portion of the slide 540 or slide pair 543 to include the sample.

A horizontal plate isolates the heat exchanger fins 623 6 from the hydration fluid supply 624. The heat DY~h~rlDr fins 623 7 may each comprise a resistive element such as a metallic wire, 8 coupled to a voltage source 634 ~;~pos~S outside the incubation 9 chamber 622. The voltage source 634 is coupled to a voltage regulator 635 to regulate the temperature of the incubation ll chamber 622, and thus of the slides 540 or slide pairs 543, to a 12 selected temperature in steps of 1 degree Celsius between ambient 13 f ~LUL~ to about 100 degrees Celsius. Heating elements and 14 regulators are known in the art.

16 The incubation oven 620 is triggered when first coupled 17 to the robotic system, and controlled to a temperature selected 18 by the control station 14. Typically, the control station 14 19 wi~l set the regulated t~ aLuLe of the incubation oven 620 to a room t ~tUL~ such as 25 degrees Celsius, will set the 21 regulated , ~LUL~ of the incubation oven 620 to an operating 22 t ~LuL~ such as 95 degrees Celsius a few minutes before the 23 incubation oven 620 is to be used in a process stQp, and will set 24 the regulated temperature of the incubation oven 620 to a room temperature or to a second operating t~ ~ ~tULe such as 37 26 degrees Celsius after the incubation oven 623 is used in a 27 process step and before it is to be used in a second process Wo~sl33240 2 1 q 1 7 7 6) PCT~S~5~06l~6 1 step. Each process step designating the incubation oven 620 2 indicates an operating temperature for that process step.

4 The hydration fluid supply 624 comprises a source, such as a bottle, into which a hydrating fluid 636 is placed and from 6 which hydrating fluid 636 is drawn during operation of the 7 incubation oven 620, and a fluid well 637 in which a selected 8 ievel of hydrating fluid 636 is maintained. The selected level 9 of hydrating fluid 636 is maintained by means of an automatic replenisher having a combination of a reservoir and valve, 11 ~icposed to maintain a constant level of hydrating fluid 636 in 12 the fluid well 637 available for evaporation into the incubation 13 chamber 622, similar to a bird feeder. The fill/drain control 14 626 provides for filling and draining the hydrating fluid 636 from the fluid well 637. Flow regulation and fluid level 16 regulation are known in the art.

18 The selected level of hydrating fluid 636 may be 19 ad~usted to account for differing assay protocols. For example, an assay protocol for hybridization may generally require heating 21 and cooling without drying out the sample. Alternatively, other 22 assay protocols, such as those for heating a xylene mixture, may 23 require a relatively dry heat.

In a preferred '~ , the hydrating fluid 636 may 26 comprise (per 10 liters) 9980 milliliters nanopure water, 20 27 milliliters Tween-20, and lo grams sorbic acid. However, those 28 skilled in the art would recognize, after perusal of this W095133240 ~ 7 7 6 P ~ . . IS6 1 application that plain water/ a known buffer solution, or another 2 substance for incubation of tissue, would also be workable for 3 the hydrating fluid 636, and that such substances would be within 4 the scope and spirit of the invention.

6 The internal cooling element 625 is disp~c~ in the 7 chassis 621 near the incubation chamber 622 to cool the 8 incubation oven 620 and those of its elements that do not need to 9 have an raised temperature. The internal cooling element 625 comprises a fan 638 coupled to the voltage source 634 and to a 11 temperature regulator 639, such as a thermostat, to ~int lin the 12 chassis 621 at a selected temperature. The heat fins 629 also 13 serve to aid in regulating the incubation chamber 622 to a 14 selected t~ a~uLe. Temperature regulation is known in the art.

17 The fluid waste receiver 627 comprises a chamber for 18 receiving excess hydrating fluid 636 not evaporated by the heat 19 exchanger fins 623, and other fluids that may be c~n~nspd by the internal cooling element 625. The fluid waste receiver 627 may 21 be detachable for emptying.

23 The receiving element 628 comprises a set of receiving 24 slots 640 molded into a bottom 641 of the incubation chamber 622, ~isposod to receive a set of feet 641 of the carrying medium 630.
26 The carrying medium's feet 641 are similar to those of the first 27 standardized slide carrier 560 or the second standardized slide W095/332~ 2 1 9 1 7 7 6 r~ IS~

1 carrier 600, so the receiving element 628 is similar to the top 2 indentations 310 of the tile 301.

4 The carrying medium 630 for inserting slides 540 or slide pairs 543 into the incubation oven 620 is similar to the 6 first standardized slide carrier 560, and comprises a frame 651, 7 a coupling ring 652, a set of slide frames 653, and a set of feet 8 654. It further comprises a slide holder cover 655, a set of 9 ventilation op~ningR 656, and a cover latch 657.

11 The frame 651 is similar to the first standardized 12 slide carrier's frame 561, and comprises a metal frame comprising 13 a set of four horizontal elements (a top 658, a slide top 659, a 14 slide bottom 660, and a bottom 661), and a set of four support posts 662. Rather than being flat as in the first standardized 16 slide carrier's frame 561, the bottom 661 comprises a V shape 17 with the bottom of the V shape in the center, to carry 18 con~l~n~tion away from the slides 540 or slide pairs 543. Other 19 frame elements may also be bent at angles or into V shapes to direct condensation away from the slides 540 or slide pairs 543.

22 The slide holder cover 655 is ~-cposed over the frame 23 651, and comprises a solid shell of a lightweight material such 24 as a rigid plastic. The slide holder cover 655 comprises a set of four shell sides 662, a set of downward sloping corners 663, 26 and a rounded topmost part 664, with the set of ventilation 27 op~n;ngc 656 defined by gaps in the topmost part 664.

W09~33240 ~ - 2~ 1 9 l 7 ~ 6 r.~ 156 1 The ventilation oponings 656 comprise a set of openings 2 665 with a slidable disk 666 dicpos~d around the coupling ring 3 652 (and related assembly) similar to the first standardized 4 slide carrier's coupling ring 562 (and related assembly, such as the ring base 571, ring bumper 573, plastic disk 574, circular 6 hole 575, circular hole 576, circular raised lip 577, and 7 circular flat portion 578). The slidable disk 666 defines a set 8 of slidable opon;ngc 667 generally CULL-~nd;ng to the 9 ventilation op~n;ng~ 656, a set of slidable masks 668 also generally C~LL c~on~l;ng to the ventilation opPnings 656, and a 11 lip 669 for sliding the slidable disk 666 to adjust the 12 ventilation opPn; ng~ 656 by alternatively uncovering them with 13 the slidable orP~;ngc 667 or covering them with the slidable 14 masks 668.

16 The slide holder cover 655 and the ventilation openings 17 656 are preferably shaped (as shown in figure 3D) to optimize 18 effects of c~n~Pn~ation of the hydratinq fluid 636 and carry 19 ron~nqate away from the slides 540 or slide pairs 543. In particular, the slide holder cover 655 and the ventilation 21 oponingc 656 are preferably trapezoidally shaped to cause the 22 hydrating fluid 636 to condense and drip back lnto the incubating 23 chamber 622, rather than evaporate into the local ai -sphpre.

The cover latch 657 comprises a ~-shaped ele~ent 669 26 coupled to one of the shell sides 662, and a peg 670 coupled to 27 the slide top 659. The V-shaped element 669 is disposed to just ~ Wo~/33~0 ~ 2 1~:9 1 7 7 6 P~Y~lSgC106l56 1 fit over the peg 670, so that a rpAcon~hly firm, but still easily 2 removable, latch is made.

4 In a preferred P~ho~ t, the incubation oven 620 is prepared with the following steps:

7 1. The operator fills the hydration fluid supply 624 8 and, if npcpcslry~ empties the fluid waste receiver 627.

2. The operator adjusts the fill/drain control 626 to 11 regulate the level of hydrating fluid 636 to a selected level.

13 3. The operator prepares the slides 540 or slide 14 pairs 543 according to a desired assay protocol, and configures the robotic system to perform the program for that assay 16 protocol.

18 4. The operator inserts the slides 540 or slide pairs 19 543 into the carrying medium 630 by means of the slide holder cover 655, and replaces the slide holder cover 655 on the 21 carrying medium 630. The operator sets the ventilation opPn; n~c 22 656 to adjust for ambient humidity levels. Preferably, the 23 vsntilation openings 656 should be as wide open as possible while 24 at the same time allowing the chemistry in the capillary gap of the slide pair 543 to maintain a level above 75% of capillary gap 26 for an entire hybridization process step.

WO !)~/33240 2~91716 ~
1 5. The operator places the slide carrying medium 630 2 in a HO~E position tile 301 and directs the control station 14 to 3 initiate the assay protocol.
The incubation oven 620 may be used in conjunction with 6 inventions disclosed in one or more of the following U.s.
7 Patents, hereby incorporated by reference as if fully set forth 8 herein: 4,731,335; 4,777,020; 4,798,706; 4,801,431; 4,975,250;
9 5,002,736; 5,023,187; and 5,116,727.
11 In a preferred ~ho~ , where the wor~station 13 12 comprises a device that should be engaged to operate on the 13 sample, coupling the carrying medium to the device requires two 14 steps: (1) The carrying medium is first coupled to or inserted lnto the device. ~2~ The device is triggered.

17 As with the incubation oven 620, the device may be 18 triggered when first coupled to the system, and controlled by the 19 control station 14. Alternatively, the device may be triggered by a switch ~triggered by contact with the robotic arm), or 21 preferably, by contact with the carrying medium by means of a 22 contact switch, proximity switch, or a weight-triggered switch 23 that detects the presence of the carrying medium or its having 24 been coupled to the device.

- =
WO95/3324U ~ 2191 7 76 r ~ 5~l56 3 Figure 3E is a flowchart of a preferred method of 4 operating the rohotic system with standardized pa~k~g~s and contents.

7 In a preferred ~ho~ir- t, at a step 681, the tray 406 8 is filled with contents 542 comprising a selected amount of a 9 selected reagent, other bioactive or chemoactive _ _ ' or mixture, or buffer.

12 At a step 682, the tray 406 has the cover 407 sealed 13 thereon.

At a step 683, the tray 406, contents 542, and cover 16 407, are transported to a location having the robotic device 10.
17 The configuration of the well dividers 522 permits the liquid 18 contents to flow easily between the inside wells 521 during 19 shi L and prior to pl~ t in a tile 301.

21 In a preferred ~hoAi- L, the contents 542 of the tray 22 406 comprise one of a set of standardized sel~to~ reagents, 23 other bioactive or chemoactive - _u--d5 or mixtures, or buffers, 24 known to p~U~L ~ of the robotic device 1o. Because the contents 542 are standardized and known to pLOyL ~ of the 26 robotic device 10, an assay protocol may be p~LUU,L -~ and 27 preloaded into the robotic device 10, for dynamic selection by an 28 operator.

W095l33240 ~ 7 7 6 PCT~Ss~6156 1 At a step 684, an operator of the robotic device 10 2 places a plurality of tiles 301 in the robotic device 10, and 3 affixes those tiles 301 to the robotic device 10 with screws or 4 other affixing objects.

6 At a step 685, the operator places one or more trays 7 406, each with its cover 407 still sealed, in a set of selected 8 tiles 301.

At a step 686, the operator removes the covers 407 from 11 the trays 406, instructs the robotic device 10 as to the location 12 of each such tray 406 and its contents 542, and ~3C the 13 robotic device lo to beqin one or more p~ U~L ' assay 14 protocols. As described herein, the pr~LU~L -3 assay protocols may be one or more assay protocols with which the 16 robotic device 10 is started, or may be one or more assay 17 protocols that are added to an already ongoing set of assay 18 protocols.

In a preferred o~ho3; t, the strength of the fixative 21 that affixes the cover 407 to the tray 406 exceeds any likely 22 force for removal that might occur during Chi, t, but is less 23 than a force for removal reguired for overcoming the spring lock 24 405. ~he U~L~t~L may therefore remove the cover 407 from the tray 406 while the tray 406 is locked into the tile 301 by means 26 of the lever 404 and the spring lock 405, without the tray 406 27 coming undone from the tile 301 due to the force of removal.

WO95/33241) l~ 2:1 :9 1 7 7 6 . ~ "~ 156 1 In a preferred ~ho~ t, the robotic device 10 2 comprises a memory with a set of p~ e~L VyL . - d assay protocols, 3 that have been previously p~OyL -~ and loaded into memory, and 4 that are selectable by a set of assay protocol names. The operator may therefore select an assay protocol by name at the 6 time it is desired to conduct the assay, without having to 7 ~epLo~L~iu the robotic device 10 each time it is desired to 8 conduct that assay. In a preferred r~=ho~;- -L, a set of 9 ~L~L~yL -' assay protocols are previously pLU~L ', transferred to an intermediate storage medium such as a diskette, ll tape, or network, and loaded into the memory of the robotic 12 device 10 by means of a operator command. The operator command 13 to load the ~L~LO~L ' protocol may also be subject to 14 security confirmation.

16 The standardized contents 542 of the trays 406 may 17 comprise a set of alcohols.

19 The standardized contents 542 of the trays 406 may comprise a set of antibodies.

22 The standardized contents 542 of the trays 406 may 23 comprise a set of hlor~inrJ agents, such as hydLugel, peroxide 24 block or a serum block.

26 The standardized contents 542 of the trays 406 may 27 comprise a set of buffer solutions, preferably a phosphate 28 buffered saline with a p~ of about 7.2. In a preferred W095/33240 2 1 9 1 7 7 6 . ~ l56 1 ~mhoA~ n~, buffer solutions should include a surfactant for best 2 operation with the capillary gap of the slide pair 542. The 3 surfactant is bridge or preferably tween ~the latter available 4 from Fisher Scientific Co.), optimized for use with the capillary qap in a slide pair 543 with about a 1% to 2~ solution of tween 6 in water.

8 The standardized contents 542 of the trays 406 may 9 comprise a set of ~11L~ ~f--, inrlll~illr those that relate to the visible range or another range of the ele~ ay~.etic ~e~Lulu 11 (such as infrared or ultraviolet).

13 The standardized contents 542 of the trays 406 may 14 comprise a set of DNA probes.

16 The standardized contents 542 of the trays 406 may 17 comprise a set of enzymes.

19 The standardized contents 542 of the trays 406 may comprise a set of fixatives.

22 The standardized contents 542 of the trays 406 may 23 comprise a set of linking molecules, such as avidin biotin 24 conjugate.

26 The standardized contents 542 of the trays 406 may 27 comprise a set of staining agents, such as hematoxylin stain or 28 eosin stain.

woss/3324o 2 1 q 1 7 7 6 ~ 6 1 The standardized contents 542 of the trays 406 may 2 comprise a set of washes, such as water.

4 A set of preferred assay protocols is described in an 5 7rpon~iY, 9 In order to use the system of this invention the operator (which might be a human user or a control pLocessuL) may 11 first determine the pL ucesses that are to be carried out the 12 apparatus. Bach step of each process may be defined. To assist 13 the user an index of work stations may be provided to allow the 14 user to determine which process steps can be employed.
Alternatively, each work station can be represented by an icon on 16 the CRT display and a help index made available that the user may 17 determine the capabilities of each work station by referring to 18 the icon and its associated help screen.

As previously described with reference to figures 1-2, 21 the ~pp~L~tUs of the invention uses a locating grid or template 22 presenting the operational work area rP~h~hlP by the robotic 23 device 10 in which the work station locations may be defined.
24 Each position on the grid is accurately determined and can be imparted to the computer to provide certainty of location. The 26 exact relative position of each work station may be stored in the 27 control system. The use of the predetermined grid locations 28 permits the user of this system to have the freedom of designing W0~5l33240 ~ 2 1 9 1 7 7 6 r~ 156 1 individual templates to match the user's need and to design the 2 steps of a process to provide relative limited ability in 3 creating yroce~s, limited only by the available work stations.

A graphic replica of the grid in which the work 6 stations located is provided on the screen of the computer, such 7 as shown in figures 6-8. Tnr~ in this graphic is the robotic 8 arm position. In order to quickly input the steps of a process to the computer (1~ a template builder and (2) a process builder lo have been created to interact with graphic replica of the work 11 area. These two tools, template builder and process builder, 12 allow the user to design a new process or modify an old process, 13 easily and quickly without the need to have knowledge of computer 14 ~ L ~.ing. Through the use of a keyboard or mouse, the two builder tools are rendered interactive with the user.

17 A work station grid area may have holes di~prced on one 18 inch centers, or any other predet~i nr d pattern. The columns of 19 holes may be identified by letters while the rows of locating holes may be identified by numbers. Thus each hole can be 21 uniquely identified by a letter-number combination.

23 Work station units or peripherals have been d~ciqnr~d 24 which have elements which cooperate with the grid locating holes and thus facilitate the exact location of each station. When 26 located on the grid each work station will have a unique 27 describer positively identifying its location.

W0~sl33240 ~ 7 7 ~ 156 1 Thus the user may _ -~rP operating the system by 2 viewing a graphic le~lesenLation of the work area surrounded by 3 icons ~S~s~ ing various work stations. As will be described 4 below the user can quickly design a new template if so desired.
Alternatively, the template may be called up from a disk by the 6 computer.

8 The steps of the process are communicated to the 9 computer through the use of an interactive peripheral such as a mouse. The operator locates the mouse cursor on the icon 11 representing the first step of the process and drags the icon to 12 the desired location. Thus by pointing and clicking the mouse 13 the work stations nec~cc~ry to accomplish the steps of the 14 process are ~icpOS~ on the graphic grid. It is of course desirable that the physical workstations be located on the grid 16 in the locations shown on the display. Alternatively, the 17 location of the work station can be fed into the computer in 18 other ways, such as through the keyboard or even by locating the 19 physical work station on the grid with feedback to the computer identifying the work station and location.

22 Thus an unsophisticated user has the ability to design 23 processes quickly imparting great flexibility to this apparatus.
24 It should of course be recognized that this information can be stored on a disk and the apparatus set up accomplished by reading 26 the information off a disk into the memory of the computer.

~og~/33~0 ~1 q 1 7 7 6 r ~ s~

1 In creating the template the operator uses a mouse to 2 draw replicas of each station on the screen, such as shown in 3 figure 7, a template building screen. Each station is given a 4 unique identification which may be a name, symbol or code. The ~i ~ionR of the station may be drawn on the screen and in 6 particular it is essential that the height of the work station is 7 recorded. The position, identificationl height and other 8 ~ n~l criteria are stored in the RAM memory of the computer 9 CPU. When the template is completed it may be stored to disk as a template file, to be recalled as needed.

12 As is not unusual in the operation of computers, 13 provisions are made to add, delete, move, resize or duplicate any 14 of the stations. Any available template previously stored may be recalled to be used or to assist in the creation of new 16 templates. Of course the apparatus may have the ability to 17 enable the operator to print out a graphic replica of the screen 18 and a list of station positions, identifications, heights or 19 other dimensions.
21 Once the template is complete the operator may use the 22 stations of the template to create a process, step by step.

24 The process builder, like the template builder, uses a graphic replica of the workstation area on the computer screen, 26 such as shown in figure 8, a process building screen. One of the 27 templates previously created by the template tool builder 28 described above, is recalled fram memory and displayed on the W095/33240 - 21 91 776 r.~ c~.

1 screen together with the work area. The screen cursor is moved 2 to the desired station icon and the particular station is 3 selected. This ~uced~Le may utilize a mouse and a point and 4 click ~L UC~dUL ~ .

- 6 Each station of the process is selected in sequence and 7 the station is then added to a list denoting the steps of the 8 process in sequential order. The robotic device would ultimately 9 be controlled to move to each of these stations in the order in which they were added the process list. Since the 11 characteristics of each work station were previously stored in 12 the computer, the robotic device would be ~IO~L ~' for the 13 proper - ~ L. For example, the height of each station was 14 previously stored in the memory, and if the robotic arm were to traverse the area in which a high work station was located, it 16 would be instructed to elevate the hand so that any sample 17 mounted thereon would clear the high work station. It is also 18 p~'~ihl~ to design the operational area to have clear paths or 19 lanes defining travel routes for the robotic device 10. In any event, the ~. L of the robotic device among the workstations 21 may be designed to be free of collisions based upon recognition 22 of the entity, position and ge~ L~y of the work stations. As 23 will appreciated as the number of work stations increase the 24 amount of information that should be considered in order to avoid collisions and otherwise avoid conflicts in instructions also 26 lncreases.

~Vogsl332~0 2 1 q l 776 . ~

1 Following the graphic design of the steps of the 2 process, the process list would be called up on the screen and 3 the pLuceduLe for each step would be imparted, such as shown in 4 figure 5. This ~L~ceduL~ would essentially indicate a range of time each sample should remain at each station. For each step a 6 minimum time and a maximum time for the sample to remain at the 7 work station would be lecoLded. As noted herein, the minimum 8 time may be specified to be zero, and the maximum time may be 9 specified to be infinity. The times for each station, except where the timing is critical, would allow the ~ystem a timing 11 window which can be used to avoid timing conflicts between 12 different steps of separate tasks and thus maximize the 13 multitasking ~p~hilities of the apparatus.

Ps~u~OCOD~ FOR DESIGNING OR RUNNING NEW PROCESSES

17 The method carried out by the control station 14 for 18 template k~ ;ng and process building may be described by 19 pcp~ ncodo shown in Tables 2-3 herein, respectively. It would be clear to one of ordinary skill in the art, after perusal of the 21 specification, drawings and claims herein, that modification of 22 known ~locessuL systems to perform the functions disclosed in 23 this ps~ nco~o (as well as in other pCP~ n.~1~ disclosed herein) 24 would be a straightforward task and would not require undue experimentation.

-21 q 1 77~
W 095/33240 ~ ,c6 procedure template tool~;
set up ~creen;
3 draw robot replica graphic;
draw grid;
display mouse cur~or;
4 sel-ct t-mplate design tool;
while (not finished) ~elect tool;
- 6 case ledit tool) add draw new statioD on screen via mouse by 7 dragging mouse away from start point while having mouse button l depressed;
8 update 5creen with a rectangle being displayed along cur50r ~ i ~r enter id Vhiaightyof 5tation;
store position and id;
select move cursor to station via mouse;
11 click mouse to select;
selected station changes color to show it is selected;

delete click mouse button l to delete;

move place move crosshair on selected station;
place cur~or on crosshair;
14 press mouse button l down and drag station to new posltion ~cr-en update aLter each new grid po~ition move;
16 resize: place resize cro~shair on selected 17 place cursor on crosshair;
press mou-e button l down and drag station to 18 new ~ize;
screen updato after each new size;
19 duplicate get curr-nt selocted station position, size ~nd height ilL i nn;
offset duplicato to new position;
add id;
21 store new ~tation position and id;

24 After the station sequence has been entered and the times for each step recorded, the process may be stored to disk 26 as a process file. The process file may be loaded in the future 27 and the apparatus used to run the same process at a later date.
28 Of course the template file may be linked to the process file so W09~33Z4~) 2 1 9 1 7 7 6 P~ S95~)615~
.

procedar- process tool~);

et up scr-en;
draw srLd;
draw proce5s li5tr 4 di~play mou~e cur~or;
case ~~ile tool) g-t t-mplat- display list of templat- fll-s;
6 ~elect Yia moqse cursor;
open ~eleoted templ-te;
7 di~play t-mplate ~tatlon~ on ~or-en;
hold station record in Ra~;
8 get process display list of process files;
s-l-ct via mous- cursor;
9 open s-lect-d proc-ss;
display proces- list in list window display associate templat- ~tation~ on the scr-en;
11 hold proces~ station records in R~M;
save process display list of proc-ss files;
12 s-lect via cursor or ~nter n-w name via keyboard;
13 stor proc--s ~ile to dluk;
14 case (iile tool~ end;
ca~e (select toolj ii cursor in work station area and on a station and mouse 16 button 1 down then add ~tation to proc-ss liat;
lf cursor in proces3 list and on list member and mou-e 17 button 1 down then del-te irom list;
18 case (select-tool) end;
case ~window ~elect~
Process i;ist: (1] ~et up screen;

display proces- in li~t mod-;
~nter min~max time via keyooard;
~croll down ~cr--n;
21 , do steps 3-4 untLl ~iniah-d;
22 6 ~xit back to pr-Vious window;
Run/Control r-turn to Run/Control window;
23 end (process toolj;

that when a process is called up from storage and run on the , ~r the template files used in the process may be auto~atically called up and displayed on the computer screen.

s WOgS~33240 ~; '=' ~2 1 9~1~7 76 r~ a 1 The procedure list on which the times at each step were 2 recorded may be called up at any time and for the stations still 3 not used by the robotic device, adjustments to the timing could 4 be made provided that the steps in the process which are to have their timing altered have not been reached. Thus the operator 6 can adjust the timing of the steps even as the process is 7 running.

g VISUAL OPERATOR INTERFACE

11 Pigure 6 shows a multitask monitoring screen 61 as 12 viewed by an operator. A multitask monitoring screen 61 may be 13 shown on a display device coupled to the computer 15, such as a 14 display monitor. The multitask monitoring screen 61 may comprise a display section 62, a menu section 63, and a status section 64.

17 The display section 62 may show a representation of the 18 robotic device 10, bench top 11, holes 12, work modules 13, and 19 related equipment. For example, the display section 62 may show positions for workstations 13 for a selected process.

22 The menu section 63 may show command options and 23 suboptions which are available to the operator and may allow the 24 operator to select one or more command options and suboptions.
For example, the menu section 63 may have a menu with the command 26 options "GET PROCESS", "BUILD PROCESS", "PROCESS LIST", "GET
27 TEMPLATE" and "BUILD TE~PLATE". The operator may display 28 available command options and select one or more command options Wogs~33240 ~ ~1 91776 r ~" ~'~'1Cf, 1 in the menu section 63, by means of a pointing device, such as a 2 mouse, as i8 well known in the art.

4 The statug section 64 may show a set of status information about processes. For example, the status secti2n 64 6 may show five pIo~esses which are in p~L~SS, and may show ~or 7 each process the current 6tep it is on, the total time it has 8 taken (both for the current step and for the cntire process), and 9 the time L- ining that it will take (both for the current step and for the entire process). Note that elapsed time for the 11 current step may be zero because the robotic device 11 might wait 12 for the proper time before depositing the sample in the 13 ~Lk~tion 13 for that process step, e.g., holding the sample in 14 the robotic hand 23 if travel from a prior step took less time than expected. The status section 64 may also show the X, Y and 16 Z position of the robotic arm.

18 Figure 7 shows a template building screen 71 as viewed 19 by an operator. A template buildin~ screen 71 may be shown on a display device coupled to the computer 15, such as a display 21 monitor, in like manner as the multitask monitoring screen 61.
22 The template b~ ing screen 71 may comprise a display section 23 62, a menu section 63, and a status section 64, in like manner as 24 the multitask monitoring screen 61.
26 When using the template building tool, described 27 herein, the operator may view the template building screen 71 and 28 ~-niplllate the -nAc and elements thereon hy means of a W09513324~ 191776 r~l~u~ 156 1 pointing device, such as a mouse. A detailed description of how 2 the operator may use the template builder tool is given herein.

4 Figure 8 shows a process building screen 81 as viewed by an operator. A process building screen 81 may be shown on a 6 display device coupled to the computer 15, such as a display 7 monitor, in like manner as the multitask monitoring screen 61.
8 The process building screen 71 may comprise a display section 62, 9 a menu section 63, and a status section 64, in liXe manner as the multitask monitoring screen 61, and a workstation section 85.

12 The workstation section 85 may show a set of names or 13 other identifiers of workstations 13. The operator may select 14 one or more w~Lk~L~tions 13 for inclusion in a process, by means of a pointing device, such as a mouse.

17 When using the process b~ ing tool, described herein, 18 the operator may view the process b~ ing screen 81 and 19 r-n i r 1 ~te the - and elements thereon by means of a 2C pointing device, such as a mouse. A detailed description of how 21 the operator may use the process builder tool is given herein.

23 Figure 9 shows a process timing screen 91 as viewed by 24 an operator. A process timing screen 91 may be shown on a display device coupled to the computer 15, such as a display 26 monitor, in like manner as the multitask monitoring screen 61.
27 The process timing screen 91 may comprise a plurality of lines 28 92, each of which may have an identifier section 93, a W~ 95/332~0 ~ ? 1 q 1 7 7 ~ , ~ 156 .

1 nameldescriptor section 94, a minimum time section 95 and a 2 maximum time section 96.

4 When usin~ the process b~ tool, described herein, the operator may view the process timing screen 91 and enter 6 minimum times (in the minimum time section 95) and maximum times 7 (in the maximum time section 96) for each process step at each 8 line 92. Each process step may thus have a line 92 with an 9 identifier in the identifier section 93 and a name or descriptor in the name/descriptor section 94.

12 The minimum time section 95 for a line 92 may specify a 13 minimu~ time which the designated process step may take, which 14 might be zero. If the minimum time is zero, additional data may be noted to indicate whether the designated process step may take 16 a single tick of a timing clock for the robotic device 10, or if 17 the designated process step may be skipped entirely.

19 The maximum time section 96 for a line 92 may speci~y a maximum time which the designated process step may take, which 21 might be infinity. If the maximum time is infinity, the system 22 may delay completion of the designated process step until after 23 all other process steps with finite maximum time have been 24 completed.
26 Each line 92 may also have an additional data section 27 97 for the designated process step, which may specify whether (1) 28 the step is to be done, (2) the step is to be skipped, or (3) the W095133240 ~ 1 9 1 776 F~~ ,i,.'01156 .

1 process is to be "held" or temporarily halted at the designated 2 process step for input from the operator. In the latter case, 3 for example, the process might be "held" at the designated 4 process step until an operator confirms that the process should 5 continue.
~ 6 9 Having delineated all the steps of all the p~ocedu~s, the computer may determine the most efficient manner for carrying 11 out the procedure. The task would be simple if the steps of the 12 first process were to be completed before the apparatus started 13 on the second process. Through the use of time interleaving, 14 multiplexing or multitasking the computer is utilized to keep track of multiple operations so as to perform a number of 16 different processes each having a multiplicity of steps 17 simultaneously.

19 In multitasking, a number of samples, each undergoing separate a~o~uL~s may all be worked on simultaneously. In time 21 interleaving, the robotic arm may operate through a sequence 22 which is determined by the timing of the individual steps of many 23 processes and the robotic arm transports different samples in a 24 time efficient se~u~nce rather than a process ordered sequence.
Although the robotic device can only move one sample to a work 26 station at a time, the entire system is continuously monitoring, 27 srhDdl~l ing and processing all tasks and their times at each 28 station ~ ur,ently. At each step the prccess performed at that ~'095'332J~ 2 1 9 1 7 7 6 ~ l5~

1 workstation continues te.g., chemical reactions) even when the 2 robotic arm is not currently attending to it. In other words, 3 the sample is ~;~pos~d in the workstation and the robotic arm 4 continues to grasp another sample. The process step continues to work on the first sa~ple while the robotic arm is attending or 6 transporting the 8econd sample. The multiple process steps that 7 are being done, one to each sample, are being done in parallel 8 and are not serial processes.

In fact the robotic arm works on a sample for a short 11 period of time durlng which it usually transports a sample to a 12 work station and then leaves that sample and works on another 13 sample or samples before returning again to the first sample.
14 Thus the robotic device work on each sample is ~n~r~r~od during the time interval that it i5 working on another sample or during 16 which the samples are being processed at a work stat$on.

18 The multitasking of the different processes is 19 A~r~ nt upon the instructions issued to the robotio device, relative to the timing of each of the 6teps in the multiple 21 ~Lucesses and the optimization of the multitasking operation~, to 22 move the samples at the s~h~ led times determined by the 23 computer inputs.

The computer control (software) may first determine all 26 the robotic ~ R necessary to complete the entire run of all 27 the steps in all the processes to be run. This determination may 28 be completed before any movement is initiated. If at any time ~ W095l33~0 - ~ 2~ 91 776 r~ 156 1 during the running of the multitasking any steps are added to one 2 or more of the processes or any of the steps are reconfigured 3 during the run, a new determination may be completed wherein the 4 ~ Dr r~rllc~lates all the v~ Ls n~rDcc~ry to complete the S run and insures that there is no time interference created by the 6 modification to the run. This method o~ predetermining the 7 m v. ts can of course be replaced by a real time method of 8 de~or~ining VG L but it is believed that the predetermining 9 method is more advantageous. The predetermining method identifies time conflicts, if any, where the robotic device would 11 be required to perform two tasks simultaneously, resolves any 12 such conflicts that may exist, and optimizes the schedule for the 13 minimum time required to complete the entire run of the multiple 14 processes.
16 This method of predetermination employs certain 17 ~r;ci~n making pLvvGduLGs which are designed to permit the 18 _~eI to resolve time conflicts and iteratively optimize the 19 srh~ le. An iterative optimization method is used because the complexity of 5rhD~Ill ing different multiple tasks, each with the 21 possibility of having multiple critically timed steps, is too 22 complex to be solved by using mathematical techniques. In 23 addition, the decision making rules allow the resolution of other 24 conflicting requirements for other resources such as the peripheral equipment or work station modules, which may be used 26 in conjunction with the robotic equipment.

W09s/33240 2 ~ 9 ~ 7 7 ~ r~ c~ 156 1 As described above, a predetermined schedule may be 2 developed to resolve time and le~UL~ conflicts and the s~h~d 3 may 4e iteratively optimized to minimi70 the time re~uired to 4 complete the steps of the multiple processes. In order to interleave the steps of the multiple processes each step of each 6 task is P~min~d at predetermined intervals, e.g., one minute. A
7 calculation is made of the time to completion of the current 8 stsp. If the step incubation time is finished a move condition 9 results. If that is the only move condition during this time, i.e., only one move condition occurs, the robotic device will be 11 scheduled to move to the next step in accordance with the 12 predet~m;nP~ schedule. However, if more than one sample is 13 scheduled to move time arbitration ensues. Time arbitration 14 de~PrminFc the fuzzy time window for each o~ the time conflicting steps and selects the sample in the most time critical step to 16 move. If more than one step has a critical time, the ~r 17 compares the times during the previous movement and varies the 18 timing of the previous tasks to resolve or prevent bottlenecks 19 from occurring. In a similar manner a single res~u~e can be srh~dnled to work on two different samples dur h g the same time 21 period and such conflicts can be resolved in a similar manner 22 using the arbitration method.

24 P5ET~OrODE FOR MUL$I~ASKING
26 The ~ethod carried out by the control station 14 for 27 multitasking may be described by ps~n~oco~P shown in Tables 4-8 28 herein. It would be clear to one of ordinary skill in the art, W095,'33240 ~ '2 1 9 1 7 7 6 PCT~S95/06l56 1 after perusal of the specification, drawings and claims herein, 2 that modification of known processor systems to perform the 3 functions disclosed in this ps~ nco~ (as well as in other 4 pseudocode ~icclnc~d herein~ would be a straight~orward task and would not require undue experimentation.
~ Tabl- 4 -- Multitasking Data Structure 8 STRUCTURE TASR ARRAY ~ 1500 element~ ]
IYTE PROCESS NUMBER;
9 BYTE TASR NUMBER;
CNAR ~25] TASK NAME;
''NTEGER TASR X COORDINATE OF WORRSTATION;
v NTEGER TASR Y COORDINATE OF WORRSTATION;
LONG INTEGER ENCODED REAL TIME FOR PICRUP OR DROPOFP;
11 C~AR tl] DROPOFF/PICRUP FLAG;
12 CHAR l5] MOVE FLAG;
~ When TRUE the prccess flagged needs to move to next task in progress. This Lnformation is entered into the 13 task array. }f multiple flags /Ire ~et ~ ne~o~ y the process steps must be arbitrated. }
14 CHAR ~5] RESOURCE FLAG;
{ If net TRUE, two or more tasks reqyire the ~me r-80urce. Re80urc~ hi~ ~,ei.~, is done to resolve all conflicts.

W095l3324u ~ i 2 1 ~9 1 7 7 6 ~ C~ 6 .

~ 5 -- 3~ultitasking ~Build Schedule) 2 PROCEDURE BUILD MULTITASK SCHEDULE () ~ Thi~ routLn i3 called a number of times with different 3 ~eeding to build a st~ti~tical sampling of a number of Bch dules The calliny routine picksi the moat optimal richedule to run ~ Tn;~ o timer and pick a proce~ for ~ir~t move For it-rative ta~k~, proces~e~ will be started in variOus order- to ~eed ta-k bullder and e-tabli-h diff-rent ~ , At ~ach 6 timcr tick ~11 proce~-s are examined to check whether lt is time to mo~e to next posiition~ If TRUE the ta~k will be ~ntered into the task array at the ~cheduled time If mor-7 than one proce~a neeodr movement at the ~ame tim r tick, tlme arbitration ensues If two or mor- processe- need tho ~ame 8 re-ource, re-ource arbitration is undergone This proc-ss contlnuen until all ta~ks in all procea~eff are ccmpl-t-TIMER ~ O;
START FIRST P~OCESS;
WHILE NOT ALL PROCESSES ST M TED DO EiEOIN
INCRE~ENT TIMER BY 1;

SET TASK MOVE FLAG

START NEXT PROCESS;
13 IF MOVE FLAG ~ 1 THEN ~IME M ~IT~nTE f~F multlple moves 4 IF TASR MOVE THEN ADD TASR TO TASR AM AY ~TASK COUNTXR3 E3lD;
~HILE NOT ALL PROCES5ES COMPLETED DO BEOIN
INCREMENT TINER ~Y 1i 16 IF ANY PROCESS NEEDS MOVEMENT THEN SET TASK MO~E FLAG;
IF ~OVE FLAG ~ 1 THEN TIME AR~I~ttE ~F multLple mov-~ ~
17 IF TASK MOVE ThEN ADD TASR ARRAY ~nsX]for resource u~e ;
END;
18 END;

2>3 W095133240 ~ 2 1 9 1 7 7 6 . PCTNS9510615G

1 T~ble 6 -- Multitasking (Time Arbitrate) 2 PRO OE DURE TIME ARBITRATE () ~ If two or more processea must be moved aimultanecusly, the 3 tlmes are arbitrated, first by examining fuzzy time range and adjusting thoae proceas taakz with fuzzy time If the cclliding prccesaea are critically timed the prcceanes pricr 4 tasks are L~_LL~n~=d tc circumvent the ccllisicn This prccedure ia called in REARRANGE ARRAY ~) }
}NTEGER FUZZY TIME~C0hP the ~ E~ maximum value }
6 BYTE CRITICAL FLAG ~ o; ~ i nii~ ~ critical flag }
7 BYTE CRIT}CAL FLAG ARRAY [S~ ~ 0, 0, 0, 0, 0 };
BEGIN
8 FOR I - l TO MAX PROCESSES
IF (PROCESS [I] MOVE FLAG SET AND FUZZY TIME [Il g FUZZY TIME COMP) T~EN BEGIN
TASK MOVE = I; ~ finds shcrtest fuzzy time }
FUZZY TIME COMP - FUZZY TIME [I~;
IF (FUZZY TIME z 0) T~EN BEGIN
11 SET CRITICAL_FLAG;
SET CRITICAL ARRAY [TASXI;
12 END;
~ If two or more processes need to move i 'i~ely a 13 L~LL , ' of earlier interleaved taaks occurs to aettle conflicts at this point if a fuzzy time range 14 aettle the ccnflict the process with the shortest fuzzy time value is set tc mcve }
IF CRITICAL FLAG > 1 T~EN REAR~ANGE ARRAY ~);
ELSE
16 ADD TASX ARRAY ~TASK MOVEI;
END;

=

W09sl3324tl ~ :21:~ ~ 7, 6 . ~ 6 q!~le 7 -- Multitasking (Resource Arbitrate 2 PROOE DURE RESOURCE ARBITRATE () I If two or more proceu~e~ neod the ~ame re~ourc- ~physlcal 3 locatLonj, fuzzy time~ for the prooe~e~ ln que~tion aro ~xamined to ~valuate whether the time slack can nettl- the 4 conflict. If not, the proce~ee prior t~uk~ are re~rranged to ri .~ th collision.
BYTE CRITICAL FLAG ~ 0; ~ Lnitlalize critlcal flag BYTE CRITICAL FLAG ARRAY [S] - ; 0, 0, 0, O, O ~;

~ Oompare prooe~a ta~k fuzzy time with other proce-~ actual 7 ta~k tLme. ~

CRITICAL PROCESS Z_TASK TIME, g IF >TAS~ MOVE = PROOE SS 2;
ELSE
COMPARE CRITICAL P~OCESS 2 FUZZY TIME WITK
CP~ITICAL PROCESS 1 TASK TIME;
IF ~TASE MOVE ~ PROCESS l;
11 IF TAS~ NOVE TRUE
ADD TASE ARRAY ITAS~ MOVEJ;
ELSE BEGIN
12 S''T CRITICAL FLAG;
S T CP~ITICAL FLAG ARRAY ITASRIi 13 R ARRANGE TAS~ ARRAY l);
END;
14 END;

Woss/3324n ~ 7 7 ~ r~
Ll ~ U

1 Table 8 -- Multitasking (Rearrange Tasks) 2 PROCEDURE PEARRANGE TASX ARRAY () 3 ~ To prevent conilict~ which cannot be arbitrated with fuzzy timing the processes in conflLct are examined at their previoua 4 step(3) and timing adju~ted in that task to remedy the conflict at the current task. After time adju~i of the crltical proce~ the taek arr~y i~ reset to the newly adjusted position and return~ to the multita3k builder and reworks the rest of the ta~ks in all processes.
BEGIN
7 I Find the la~t time the critical proc-sli WZ18 moved.
REPEAT
8 POSITION = posITIon - l;
UNTIL TASR ARRAY [POSITION~ = CRITICAL FLAG ARRAY [TASRj;
~ Adju3t tim-r. }
INCP~E~ENT TASK ~TAS~ ARRAY [POSITION].~IN TI~E] BY X;
~ Reset position and time. ~
11 SET POSITION TO CUP~RENT TASR ARRAY VALUE;
SET TI~ER TO CUPRENT TASR ARRAY VALUE;
12 PETURN TO ~5ULTITASR RUILDER;
13 END;

17 It would be clear to one of ordinary skill in the art, lô after perusal of the specification, drawings and claims herein, 19 that there is a multitude of interleave paths that can be taken to achieve multitasking of a plurality of processes. Each path 21 will in all probability have a different time to complete all of 22 the steps of all of the processes. In view of this it will be 23 appreciated that for optimum efficiency it is necessary to select 24 the optimum path which will take the minimum time to complete.
As a practical matter an iterative process can be used in which 26 the interleave path is computed several times. Eiach time the 27 interleave variables are iterated they are ordered and computed 28 differently so that different results are obtained for each ~YO9~3324(~ 9l 7 7 ~ PCT~S~106156 1 iteration. The number of iterations necessary to arrive at an 2 optimized path can be computed statistically by taking the number 3 of steps in each task and the number of taBks to be performed.
4 Since run time of the paths calculated from the uus iterations follow a normal distribution curve, the minimum number 6 of iterations noroc~ry to achieve a path that will be among the 7 ~aster run times can be calculated.

9 One technique for computing an optimal interleave path may compute a set o~ interleave paths by iterating a selected 11 number o~ times in response to the number of steps in each task 12 and the number of tasks to be performed. The number of 13 iterations may alternatively be selected to be a fixed number, 14 such as 20 iterations, that may be altered in ~ ~~e to a command from an operator.

17 In a preferred '~--nt, multiple tasks may be run 18 with disjoint workstations, since it is possible that a reagent, l9 or other chemoactive or bioactive _ ~ or mixture, at a workstation will be contaminated by the sample tissue on the 21 slide. However, where it iB believed that contamination would be 22 minimal, or at least that effects of such contamination would be 23 minimal, it would alternatively be preferable to share resources 24 such as standard buffers, washes, and pads. In this alternative embodiment, a source for a standard buffer or wash would be made 26 available by means of an automatic replenisher having a 27 combination of a reservoir and valve, disposed to maintain a wo g5,332~0 ~ 9 1 7 7 6 PCT~S95l06156 1 constant level of liquid available for dipping a slide, similar 2 to a bird feeder.

4 In an alternative ~ , it may be preferable to design protocols for multiple simultaneous tasks to use a maximum 6 set of common reagents or workstations. It would be preferable 7 to design such protocols in two parts, part 1 and part 2, 8 separated by a selected time, so that a set of ~esou. eeS used in 9 part 1 of the protocol are not used in part 2 of the protocol.
With this design, a leS~u~e arbitration technique may more 11 easily distinguish when it is possible to start a second 12 instantiation of the same protocol.

14 APPE~DIX

16 A preferred set of protocols are shown in an Arp~n~; y 17 to this specification, hereby incorporated by reference as if 18 fully set forth herein. These protocols are Copyright 1994 19 Biotek Solutions, Inc., and their inclusion in this patent application is not a waiver of copyright or any of the rights 21 afforded by copyright.

23 Each protocol is intended for operation on the TechMate 24 (TM) robotic controller (available from Biotek Solutions, Inc. of Santa sarbara, California), and includes the following sections:

27 o a protocol program name, a brief title, and an ~YrAn~d 28 title;

2 1 9 1 1 7~
W09~/33241~ 56 1 o a summary o~ the running time;

3 o a description of the principles of operation for the 4 protocol;

6 o a description of the nature of the spe~i (s) the 7 protocol is intended to operate upon;

9 o a description of the nature of the preparation for the specimen(s) the protocol is intended to operate upon;

12 o a description of the nature of the preparation for 13 chemical reagents the protocol is intended to operate 14 with;

16 o a description of the pLoceduLe used in operation of the 17 protocol;

19 o a description of the expected results from operation of the protocol;

22 o a description of references for further information 23 about the principles of operation for the protocol;

o an ordered listing of program steps; and 27 o a map template for operation of the protocol.

WO~J5/33240 2 -1 9 1 7 7 6 PCT/US~5106156 .

1 For each protocol, the ordered listing of program steps 2 comprises five columns:

4 o a sequence number, indicating a step number for the indicated program step;
7 o a protocol operation namel indicating a protocol 8 operation to be performed at the indicated step number;

o a minimum time duration, indicating a minimum duration 11 the indicated protocol operation may be performed, in 12 hours, minutes, and seconds;

14 o a maximum time duration, indicating a maximum duration the indicated protocol operation may be performed, in 16 hours, minutes, and seconds; and 18 o an indicator of whether the step is actually performed, 19 . where "Y" = yes and "N" = no; or an oven temperature may be designated.

22 The protocol operation may comprise one of the 23 following:

100% 100% ethanol 26 50% EtOH 50% ethanol 27 5~ HCL 5 normal hydrochloric acid 28 AALC absolute alcohol W09~s/33240 ~ 1 9 1 7 7 6 F~ J . '~X1~6 l ABl primary antibody -- ABlA and A~lB also 2 indicate a primary antibody 3 AB2 se~vllddLy antibody 4 ~3C avidin biotin conjugate AP alkaline phosphatase (enzyme detection) 6 BLECH bleach 7 BLOR blocking antibody, i.e., a bioactive agent 8 that blocks secondary antiho~ipc that are 9 already present in the robotic system BUFxxx a phosphate buffer, as noted herein ll CHROM GEN a chromagen 12 DAB diamino benzidine 13 ENZ an enzy~e, e.g., to help open up antigenic 14 sites EOSIN eosin 16 FK a fluorescent ~I-s gsn 17 H20 water i8 HEMA hematoxylin 19 HI WASH a high stringency (high ionic cv,lv~.. LL~tion~
wash, typically used for DNA probes 21 HOME a "home" location for starting and/or 22 stopping an assay protocol 23 HP HP block, e.g., to block enzymes that are 24 endogenous to the robotic system HYPO sodium thiosulfate, a reducing agent used to 26 remove some mercury-based fixatives 27 IO iodlne 28 IP an i -s~rum~ e.g., for enzyme detection W09s~33240 .-2 1 ~9 1 7 7 6 ~"~ rl5~

1 LO WASH a low stringency (low ionic ~n~ L~tion) 2 wash, typically used for DNA probes 3 NE BL methylene blue stain 4 PADxxx a blotter, preferably 1/2 inch thick PARK a location to wait until a next step ~ 6 PROBE a DNA probe 7 SCHIF Schiff reagent for a Schiff reaction 8 STN a stain 9 XY xylene 11 Those skilled in the art will recognize, after perusal 12 of this application, that other and further protocol operations, 13 reagents, chemoactive or bioactive ~ buffers, or other 14 substances would be workable with the devices and substances disclosed herein, and are within the scope and spirit of the 16 invention.

18 Alternative ~hn~

While preferred embodiments are disclosed herein, many 21 variations are possible which remain within the concept, scope, 22 and spirit of the invention, and these variations would become 23 clear to those skilled in the art after perusal of this 24 application.

26 For example, it would become clear to those skilled in 27 the art that the devices and techniques described herein would be 28 applicable to other processes, subject to standardization and ~og~33240 2 1 9 1 7 7 6 ~ cl~6 1 robotic operation, and that such application would be within the 2 concept, scope, and spirit of the invention. Such pLO~e~
3 could include those related to dcveloping film and those related 4 to manufacture or testing of electronic circuit6, printed circuit boards, or semiconductor wafers.

7 For a second example, it would beco~e clear to those 8 skilled in the art that the devices and techniques described 9 herein for use with liquid would generally be applicable to 10 ~LOce~Se~ using other flowable substances, including colloids, 11 gels, or powders, and that such application would be within the 12 concept, scope, and spirit of the invention.

ZO

Claims (100)

We claim:
1. A method for performing a plurality of independent analysis procedures simultaneously, each said procedure having a sample and at least one process step for operating on that sample, said method comprising selecting, at a plurality of times, a sample to be moved;
directing a robotic arm to move said sample to be moved by interleaving the process steps of said plurality of independent analysis procedures;
monitoring progress information for said procedures;
and altering a sequence of said process steps in response to said progress information and in response to information from an operator;
wherein said step of altering comprises the steps of (1) generating a possible new sequence of process steps from a time said altering occurs onward; (2) examining said possible new sequence for possible conflicts; and (3) altering said possible new sequence in response to said timing information and said possible conflicts;
wherein at least one of said process steps comprises a bioassay workstation; a biomedical workstation; a chemical process workstation; a heat process workstation; an irradiation process workstation; a centrifuge, a diffusion device, a distillation device, a separation device; a DNA crosslinking device; an electroporator; a laser device, an optical device; a microwave device, a radioactive sample, a radiation source; an incubation oven, a heating unit; a refrigeration element, a cooling unit; or a workstation in which a process is to be performed comprising at least one of the following substances: an alcohol, an antibody, an antibody carrier, an antibody probe, benzene, cellulose nitrate, chloroform, a chromophore, a colored staining solution, a colormetric substrate, a counterstain, a dehydrating fluid, a DNA probe, an enzyme labeled detection system, eosin, epoxy resin, ester wax, ethanol, a fat solvent, a fixative for electron microscopy, a fixative for optical microscopy, a fluorescent chromagen, formaldehyde, formalin, glutaraldehyde, hematoxylin, a heavy metal salt, mercuric chloride, methacrylate, an organic reagent, osmium tetroxide, a paraffin type wax, picric acid, a plastic substance, a substance on which a selected enzyme is known to have a specified effect, a synthetic dye, toluene, a washing buffer, water, a water soluble dye, a wax, or a test suited to identify a specific chemical substance or enzyme within the tissue sample.
2. A method for performing a plurality of independent analysis procedures simultaneously, each said procedure having a sample and at least one process step for operating on that sample, said method comprising selecting, at a plurality of times, a sample to be moved;

directing a robotic arm to move said sample to be moved by interleaving the process steps of said plurality of independent analysis procedures;
monitoring progress information for said procedures;
and altering a sequence of said process steps in response to said progress information and in response to information from an operator;
wherein said step of altering comprises the steps of (1) generating a possible new sequence of process steps from a time said altering occurs onward; (2) examining said possible new sequence for timing conflicts occurring before a known time value, (3) advancing said known time value from the time said altering occurs to an end of said possible new sequence, (4) selecting an exact time to start said first process step when a first process step is found to have a timing conflict with a second process step and said first process step has a range of times at which it may be started, (5) backtracking said known time value and altering said possible new sequence starting from said backtracked known time value to avoid said timing conflict, when a first process step is found to have a timing conflict with a second process step and said first and second process steps have exact times at which they may be started, and (6) signalling an error when said known time value is backtracked beyond the time said altering occurs;
wherein at least one of said process steps comprises a bioassay workstation; a biomedical workstation; a chemical process workstation; a heat process workstation; an irradiation process workstation; a centrifuge, a diffusion device, a distillation device, a separation device; a DNA crosslinking device; an electroporator; a laser device, an optical device; a microwave device, a radioactive sample, a radiation source; an incubation oven, a heating unit; a refrigeration element, a cooling unit; or a workstation in which a process is to be performed comprising at least one of the following substances: an alcohol, an antibody, an antibody carrier, an antibody probe, benzene, cellulose nitrate, chloroform, a chromophore, a colored staining solution, a colormetric substrate, a counterstain, a dehydrating fluid, a DNA probe, an enzyme labeled detection system, eosin, epoxy resin, ester wax, ethanol, a fat solvent, a fixative for electron microscopy, a fixative for optical microscopy, a fluorescent chromagen, formaldehyde, formalin, glutaraldehyde, hematoxylin, a heavy metal salt, mercuric chloride, methacrylate, an organic reagent, osmium tetroxide, a paraffin type wax, picric acid, a plastic substance, a substance on which a selected enzyme is known to have a specified effect, a synthetic dye, toluene, a washing buffer, water, a water soluble dye, a wax, or a test suited to identify a specific chemical substance or enzyme within the tissue sample.
3. A method for performing a plurality of independent analysis procedures simultaneously, each said procedure having a sample and at least one process step for operating on that sample, said method comprising the steps of selecting, at a plurality of times, a sample to be moved, said step of selecting comprising the steps of (1) generating a plurality of possible sequences of process steps, less than all possible sequences, (2) determining statistical information about a time distribution of said plurality, (3) selecting a preferred one of said plurality with a desired total required time, so as to substantially minimize a total time required to complete said procedures, and (4) selecting said sample to be moved in accordance with said preferred one of said plurality of possible sequences;
directing a robotic arm to move said sample to be moved by interleaving the process steps of said plurality of independent analysis procedures;
wherein at least one of said process steps comprises a bioassay workstation; a biomedical workstation; a chemical process workstation; a heat process workstation; an irradiation process workstation; a centrifuge, a diffusion device, a distillation device, a separation device; a DNA crosslinking device; an electroporator, a laser device, an optical device; a microwave device, a radioactive sample, a radiation source; an incubation oven, a heating unit; a refrigeration element, a cooling unit; or a workstation in which a process is to be performed comprising at least one of the following substances: an alcohol, an antibody, an antibody carrier, an antibody probe, benzene, cellulose nitrate, chloroform, a chromophore, a colored staining solution, a colormetric substrate, a counterstain, a dehydrating fluid, a DNA probe, an enzyme labeled detection system, eosin, epoxy resin, ester wax, ethanol, a fat solvent, a fixative for electron microscopy, a fixative for optical microscopy, a fluorescent chromagen, formaldehyde, formalin, glutaraldehyde, hematoxylin, a heavy metal salt, mercuric chloride, methacrylate, an organic reagent, osmium tetroxide, a paraffin type wax, picric acid, a plastic substance, a substance on which a selected enzyme is known to have a specified effect, a synthetic dye, toluene, a washing buffer, water, a water soluble dye, a wax, or a test suited to identify a specific chemical substance or enzyme within the tissue sample.
4. A method of operating a processor having a display screen for specifying a test procedure in a system for performing a plurality of simultaneous test procedures, said method comprising the steps of displaying a plurality of templates on said screen for viewing by an operator;
receiving a first signal from said operator indicative of a first location on said screen;
identifying said location with a first one of said templates;
receiving a second signal from said operator indicative of a second location on said screen;
displaying said first one of said templates at said second location;
identifying a process step with said template and said second location;
identifying a location and order for said process step to be performed in response to said second location; and repeatedly performing said steps until an ordered sequence of said process steps has been determined.
5. A method as in claim 4, comprising the step of receiving a third signal from said operator indicative of information about said process step.
6. A method of operating a procedure having a display screen for specifying a test procedure in a system for performing a plurality of test procedures, said method comprising the steps of selecting a first location on said screen within a template displayed on a screen;
moving a copy of said template to a second location on said screen;
identifying a process step with said template and said second location;
identifying a location and order for said process step to be performed in response to said second location; and repeatedly performing said steps until an ordered sequence of said process steps has been determined.
7. A method of operating a system for performing a plurality of independent analysis procedures simultaneously, each said procedure having a sample and at least one process step for operating on that sample, said method comprising coupling a plurality of tiles to a substrate, each at a selected location on said substrate;
coupling a plurality of sealed reagent trays, each having a substantially uniform shape and size selected from a predetermined set of shapes and sizes, each having been prefilled with an amount of a substance, said amount and said substance selected from a predetermined set of amounts and substances, to said plurality of tiles;
exposing said substance in said sealed reagent trays while said sealed reagent trays are coupled to said plurality of tiles;
selecting a plurality of analysis programs from a set of predetermined analysis programs stored in a computer memory;
and initiating an interleaving program stored in a computer memory that performs said plurality of analysis programs simultaneously by directing a robotic device to move said samples among said plurality of reagent trays, interleaving a plurality of steps of said analysis programs.
8. A method for performing a plurality of independent analysis procedures simultaneously, each said procedure having a sample and at least one process step for operating on that sample, said method comprising disposing said sample in a slide holder, said slide holder comprising a set of slides disposed in an array;
disposing said slide holder in an incubation chamber having a plurality of heat exchanger elements disposed in an array matched to said slides;
applying a selected amount of a hydration fluid to said sample; and applying a regulated amount of energy to said heat exchanger elements to achieve a selected temperature at said sample.
9. A method as in claim 8, comprising adjusting a set of ventilation openings coupled to said incubation chamber.
10. A method as in claim 8, comprising cooling said incubation chamber while said slides are disposed therein.
11. A method as in claim 10, wherein said step of cooling comprises operating a fan using a temperature regulator.
12. A method as in claim 8, comprising disposing a structural member near said slides to direct condensation away from said slides.
13. A method as in claim 8, wherein said step of applying a regulated amount of energy comprises cooling said incubation chamber.
14. A method as in claim 8, wherein said step of applying a selected amount of a hydration fluid comprises maintaining a level of hydration fluid in a fluid well coupled to said incubation chamber at a selected level.
15. A method as in claim 8, wherein said step of applying a selected amount of a hydration fluid comprises applying a hydration fluid having about .1% to about 3% of a surfactant.
16. A method as in claim 8, wherein said step of applying a selected amount of a hydration fluid comprises applying a hydration fluid having less than about 3% of a surfactant.
17. A system for performing a plurality of independent analysis procedures simultaneously, each said procedure having a sample and at least one process step for operating on that sample, said system comprising a robotic arm for moving the samples among a plurality of processing stations;
a processor for selecting, at a plurality of times, a sample to be moved, and for directing said robotic arm to move said sample to be moved; said processor having means for directing said robotic arm to interleave the process steps of said plurality of independent analysis procedures;
means for monitoring progress information for said procedures; and means for altering a sequence of said process steps in response to said progress information and in response to information from an operator;
wherein said means for altering comprises (l) means for generating a possible new sequence of process steps from a time said altering occurs onward; (2) means for examining said possible new sequence for possible conflicts; and (3) means for altering said possible new sequence in response to said timing information and said possible conflicts;
wherein at least one of said process steps comprises a bioassay workstation; a biomedical workstation; a chemical process workstation; a heat process workstation; an irradiation process workstation; a centrifuge, a diffusion device, a distillation device, a separation device; a DNA crosslinking device; an electroporator; a laser device, an optical device; a microwave device, a radioactive sample, a radiation source; an incubation oven, a heating unit; a refrigeration element, a cooling unit; or a workstation in which a process is to be performed comprising at least one of the following substances: an alcohol, an antibody, an antibody carrier, an antibody probe, benzene, cellulose nitrate, chloroform, a chromophore, a colored staining solution, a colormetric substrate, a counterstain, a dehydrating fluid, a DNA probe, an enzyme labeled detection system, eosin, epoxy resin, ester wax, ethanol, a fat solvent, a fixative for electron microscopy, a fixative for optical microscopy, a fluorescent chromagen, formaldehyde, formalin, glutaraldehyde, hematoxylin, a heavy metal salt, mercuric chloride, methacrylate, an organic reagent, osmium tetroxide, a paraffin type wax, picric acid, a plastic substance, a substance on which a selected enzyme is known to have a specified effect, a synthetic dye, toluene, a washing buffer, water, a water soluble dye, a wax, or a test suited to identify a specific chemical substance or enzyme within the tissue sample.
18. A system for performing a plurality of independent analysis procedures simultaneously, each said procedure having a sample and at least one process step for operating on that sample, said system comprising a robotic arm for moving the samples among a plurality of processing stations;
a processor for selecting, at a plurality of times, a sample to be moved, and for directing said robotic arm to move said sample to be moved; said processor having means for directing said robotic arm to interleave the process steps of said plurality of independent analysis procedures;
means for monitoring progress information for said procedures; and means for altering a sequence of said process steps in response to said progress information and in response to information from an operator;
wherein said means for altering comprises (1) means for generating a possible new sequence of process steps from a time said altering occurs onward; (2) means for examining said possible new sequence for timing conflicts occurring before a known time value, (3) means for advancing said known time value from the time said altering occurs to an end of said possible new sequence, (4) means, when a first process step is found to have a timing conflict with a second process step and said first process step has a range of times at which it may be started, for selecting an exact time to start said first process step, (5) means, when a first process step is found to have a timing conflict with a second process step and said first and second process steps have exact times at which they may be started, for backtracking said known time value and altering said possible new sequence starting from said backtracked known time value to avoid said timing conflict, and (6) means for signaling an error when said known time value is backtracked beyond the time said altering occurs;
wherein at least one of said process steps comprises a bioassay workstation; a biomedical workstation; a chemical process workstation; a heat process workstation; an irradiation process workstation; a centrifuge, a diffusion device, A
distillation device, a separation device; a DNA crosslinking device; an electroporator; a laser device, an optical device; a microwave device, a radioactive sample, a radiation source; an incubation oven, a heating unit; a refrigeration element, a cooling unit; or a workstation in which a process is to be performed comprising at least one of the following substances: an alcohol, an antibody, an antibody carrier, an antibody probe, benzene, cellulose nitrate, chloroform, a chromophore, a colored staining solution, a colormetric substrate, a counterstain, a dehydrating fluid, a DNA probe, an enzyme labeled detection system, eosin, epoxy resin, ester wax, ethanol, a fat solvent, a fixative for electron microscopy, a fixative for optical microscopy, a fluorescent chromagen, formaldehyde, formalin, glutaraldehyde, hematoxylin, a heavy metal salt, mercuric chloride, methacrylate, an organic reagent, osmium tetroxide, a paraffin type wax, picric acid, a plastic substance, a substance on which a selected enzyme is known to have a specified effect, a synthetic dye, toluene, a washing buffer, water, a water soluble dye, a wax, or a test suited to identify a specific chemical substance or enzyme within the tissue sample.
19. A system for performing a plurality of independent analysis procedures simultaneously, each said procedure having a sample and at least one process step for operating on that sample, said system comprising a robotic arm for moving the samples among a plurality of processing stations; and a processor for selecting, at a plurality of times, a sample to be moved, and for directing said robotic arm to move said sample to be moved; said processor having means for directing said robotic arm to interleave the process steps of said plurality of independent analysis procedures;
wherein said processor comprises a display screen showing a set of predetermined symbols representing process steps;
means for drawing one of said symbols on said display screen in response to information from an operator;
means for associating a process step with a location where said process step is to be performed; and means for associating a processing station with said one symbol;
wherein at least one of said process steps comprises a bioassay workstation; a biomedical workstation; a chemical process workstation; a heat process workstation; an irradiation process workstation; a centrifuge, a diffusion device, a distillation device, a separation device; a DNA crosslinking device; an electroporator; a laser device, an optical device; a microwave device, a radioactive sample, a radiation source; an incubation oven, a heating unit; a refrigeration element, a cooling unit; or a workstation in which a process is to be performed comprising at least one of the following substances: an alcohol, an antibody, an antibody carrier, an antibody probe, benzene, cellulose nitrate, chloroform, a chromophore, a colored staining solution, a colormetric substrate, a counterstain, a dehydrating fluid, a DNA probe, an enzyme labeled detection system, eosin, epoxy resin, ester wax, ethanol, a fat solvent, a fixative for electron microscopy, a fixative for optical microscopy, a fluorescent chromagen, formaldehyde, formalin, glutaraldehyde, hematoxylin, a heavy metal salt, mercuric chloride, methacrylate, an organic reagent, osmium tetroxide, a paraffin type wax, picric acid, a plastic substance, a substance on which a selected enzyme is known to have a specified effect, a synthetic dye, toluene, a washing buffer, water, a water soluble dye, a wax, or a test suited to identify a specific chemical substance or enzyme within the tissue sample.
20. A system for performing a plurality of independent analysis procedures simultaneously, each said procedure having a sample and at least one process step for operating on that sample, said system comprising a robotic arm for moving the samples among a plurality of processing stations; and a processor for selecting, at a plurality of times, a sample to be moved, and for directing said robotic arm to move said sample to be moved; said processor having means for directing said robotic arm to interleave the process steps of said plurality of independent analysis procedures;
wherein said processor comprises a memory for storing timing information for each said process step, said timing information comprising a predetermined range of durations during which said process step may be in a predetermined state; and means for determining an exact time to start each said process step in a first said procedure in response to timing information comprising for at least one step in a second said procedure;
wherein at least one of said process steps comprises a bioassay workstation; a biomedical workstation; a chemical process workstation; a heat process workstation; an irradiation process workstation; a centrifuge, a diffusion device, a distillation device, a separation device; a DNA crosslinking device; an electroporator; a laser device, an optical device; a microwave device, a radioactive sample, a radiation source; an incubation oven, a heating unit; a refrigeration element, a cooling unit; or a workstation in which a process is to be performed comprising at least one of the following substances: an alcohol, an antibody, an antibody carrier, an antibody probe, benzene, cellulose nitrate, chloroform, a chromophore, a colored staining solution, a colormetric substrate, a counterstain, a dehydrating fluid, a DNA probe, an enzyme labeled detection system, eosin, epoxy resin, ester wax, ethanol, a fat solvent, a fixative for electron microscopy, a fixative for optical microscopy, a fluorescent chromagen, formaldehyde, formalin, glutaraldehyde, hematoxylin, a heavy metal salt, mercuric chloride, methacrylate, an organic reagent, osmium tetroxide, a paraffin type wax, picric acid, a plastic substance, a substance on which a selected enzyme is known to have a specified effect, a synthetic dye, toluene, a washing buffer, water, a water soluble dye, a wax, or a test suited to identify a specific chemical substance or enzyme within the tissue sample.
21. A system for performing a plurality of independent analysis procedures simultaneously, each said procedure having a sample and at least one process step for operating on that sample, said system comprising a robotic arm for moving the samples among a plurality of processing stations; and a processor for selecting, at a plurality of times, a sample to be moved, and for directing said robotic arm to move said sample to be moved; said processor having means for directing said robotic arm to interleave the process steps of said plurality of independent analysis procedures;
wherein said processor comprises means for generating a plurality of possible sequences of process steps, less than all possible sequences;
means for determining statistical information about a time distribution of said plurality; and means for selecting one of said plurality with a desired total expected time, so as to substantially minimize a total time required to complete said procedures;

wherein at least one of said process steps comprises a bioassay workstation; a biomedical workstation; a chemical process workstation; a heat process workstation; an irradiation process workstation; a centrifuge, a diffusion device, a distillation device, a separation device; a DNA crosslinking device; an electroporator; a laser device, an optical device; a microwave device, a radioactive sample, a radiation source; an incubation oven, a heating unit; a refrigeration element, a cooling unit; or a workstation in which a process is to be performed comprising at least one of the following substances: an alcohol, an antibody, an antibody carrier, an antibody probe, benzene, cellulose nitrate, chloroform, a chromophore, a colored staining solution, a colormetric substrate, a counterstain, a dehydrating fluid, a DNA probe, an enzyme labeled detection system, eosin, epoxy resin, ester wax, ethanol, a fat solvent, a fixative for electron electron microscopy, a fixative for optical microscopy a fluorescent chromagen, formaldehyde, formalin, glutaraldehyde, hematoxylin, a heavy metal salt, mercuric chloride, methacrylate, an organic reagent, osmium tetroxide, a paraffin type wax, picric acid, a plastic substance, a substance on which a selected enzyme is known to have a specified effect, a synthetic dye, toluene, a washing buffer, water, a water soluble dye, a wax, or a test suited to identify a specific chemical substance or enzyme within the tissue sample.
22. A system for performing a plurality of independent analysis procedures simultaneously, each said procedure having a sample and at least one process step for operating on that sample, said system comprising a robotic arm for moving the samples among a plurality of processing stations; and a processor for selecting, at a plurality of times, a sample to be moved, and for directing said robotic arm to move said sample to be moved; said processor having means for directing said robotic arm to interleave the process steps of said plurality of independent analysis procedures;
wherein said processor comprises means for monitoring dynamic progress information for said procedures; and means for altering a sequence of said process steps in response to said progress information and in response to information from an operator;
wherein at least one of said process steps comprises a bioassay workstation; a biomedical workstation; a chemical process workstation; a heat process workstation; an irradiation process workstation; a centrifuge, a diffusion device, a distillation device, a separation device; a DNA crosslinking device; an electroporator; a laser device, an optical device; a microwave device, a radioactive sample, a radiation source; an incubation oven, a heating unit; a refrigeration element, a cooling unit; or a workstation in which a process is to be performed comprising at least one of the following substances: an alcohol, an antibody, an antibody carrier, an antibody probe, benzene, cellulose nitrate, chloroform, a chromophore, a colored staining solution, a colormetric substrate, a counterstain, a dehydratihg fluid, a DNA probe, an enzyme labeled detection system, eosin, epoxy resin, ester wax, ethanol, a fat solvent, a fixative for electron microscopy, a fixative for optical microscopy, a fluorescent chromagen, formaldehyde, formalin, glutaraldehyde, hematoxylin, a heavy metal salt, mercuric chloride, methacrylate, an organic reagent, osmium tetroxide, a paraffin type wax, picric acid, a plastic substance, a substance on which a selected enzyme is known to have a specified effect, a synthetic dye, toluene, a washing buffer, water, a water soluble dye, a wax, or a test suited to identify a specific chemical substance or enzyme within the tissue sample.
23. A system for performing a plurality of independent analysis procedures simultaneously, each said procedure having a sample and at least one process step for operating on that sample, said system comprising a robotic device for moving the samples among a plurality of processing stations; and a processor for selecting, at a plurality of times, a sample to be moved, and for directing said robotic arm to move said sample to be moved; said processor having means for directing said robotic arm to interleave the process steps of said plurality of independent analysis procedures in a sequence conforming to a predetermined range of durations for each one of said process steps.
24. A system as in claim 23, comprising a data structure having a sequence of process steps indexed by a time value and indicating a range of possible durations for each said process step.
25. A system as in claim 23, comprising a display area for specifying a test procedure;
means for selecting a first location on said display area within a template displayed thereon;
means for moving a copy of said template to a second location on said display area; and means for identifying a process step and a sequence order for said process step, in response to said template and said second location.
26. A system as in claim 23, comprising means for monitoring dynamic progress information for said plurality of independent procedures; and means for altering a sequence of said process steps in response to said progress information and in response to information from an operator.
27. A system as in claim 26, wherein said means for altering comprises means for receiving a command from said operator for changing said sequence of process steps; and means for determining a new sequence of process steps in response to said command and in response to timing information about said process steps.
28. A system as in claim 23, wherein at least one of said process steps comprises a bioassay workstation; a biomedical workstation; a chemical process workstation; a heat process workstation; an irradiation process workstation; a centrifuge, a diffusion device, a distillation device, a separation device; a DNA crosslinking device; an electroporator; a laser device, an optical device; a microwave device, a radioactive sample, a radiation source; an incubation oven, a heating unit; a refrigeration element, a cooling unit; or a workstation in which a process is to be performed comprising at least one of the following substances. an alcohol, an antibody, an antibody carrier, an antibody probe, benzene, cellulose nitrate, chloroform, a chromophore, a colored staining solution, a colormetric substrate, a counterstain, a dehydrating fluid, a DNA
probe, an enzyme labeled detection system, eosin, epoxy resin, ester wax, ethanol, a fat solvent, a fixative for electron microscopy, a fixative for optical microscopy, a fluorescent chromagen, formaldehyde, formalin, glutaraldehyde, hematoxylin, a heavy metal salt, mercuric chloride, methacrylate, an organic reagent, osmium tetroxide, a paraffin type wax, picric acid, a plastic substance, a substance on which a selected enzyme is known to have a specified effect, a synthetic dye, toluene, a washing buffer, water, a water soluble dye, a wax, or a test suited to identify a specific chemical substance or enzyme within the tissue sample.
29. A system as in claim 23, wherein said plurality of processing stations are disposed in a set of grid locations, and wherein said robotic device comprises a bench robot with sufficient degrees of freedom that it is able to reach each of said plurality of processing stations with suitable movement;
means for coupling to a sample;
means for holding a sample while moving; and means for decoupling from a sample.
30. A system as in claim 23, wherein said processor comprises a memory for storing timing information for each said process step, said timing information comprising a range of durations during which said process step may be in a predetermined state; and means for determining an exact time to start each said process step in a first said procedure in response to timing information for at least one process step in a second said procedure.
31. A system as in claim 30, wherein said means for determining comprises means for generating a possible sequence of process steps;
means for examining said possible sequence for timing conflicts occurring before a known time value;

means for advancing said known time value from a beqinning of said possible sequence to an end of said possible sequence;
means, when a first process step is found to have a timing conflict with a second process step and said first process step has a range of times at which it may be started, for selecting an exact time to start said first process step; and means, when a first process step is found to have a timing conflict with a second process step and said first and second process steps have exact times at which they may be started, for backtracking said known time value and altering said possible sequence starting from said backtracked known time value to avoid said timing conflict.
32. A system as in claim 23, wherein said processor comprises means for generating a plurality of possible sequences of process steps, less than all possible sequences;
means for determining statistical information about a time distribution of said plurality of possible sequences; and means for selecting one of said plurality of possible sequences with a desired total expected time, so as to substantially minimize a total time required to complete said procedures.
33. A system as in claim 23, wherein said robotic device comprises a substrate to which are coupled a plurality of tiles, each one of said tiles disposed at a selected location on said substrate;
a plurality of reagent trays coupled to said plurality of tiles, each one of said reagent trays having substantially identical shape and size;
a robotic arm disposed for reaching substantially all of said tiles and comprising means for coupling to and decoupling from a sample holder, said sample holder having means for holding at least one sample in a selected position, whereby said robotic arm is capable of moving said sample from a first one of said reagent trays to a second one of said reagent trays.
34. A system as in claim 33, wherein each one of said tiles comprises a plurality of receptacles, each receptacle for receiving one of said plurality of reagent trays.
35. A system as in claim 33, wherein said first one reagent tray comprises a cover disposed thereon with adhering means having sufficient power to prevent release of said cover during normal handling and shipping, but able to release said cover when said reagent tray is locked into said tile with said reagent tray lock.
36. A system as in claim 33, wherein said first one reagent tray comprises a plurality of receptacles, each for an aliquot of a substance for reaction with said sample;
a first said receptacle and an second said receptacle define a barrier disposed to prevent said substance from moving from said first said receptacle to said second said receptacle when said reagent tray is disposed in a first position, and disposed to not prevent said substance from moving from said first said receptacle to said second said receptacle when said reagent tray is disposed in a second position.
37. A system as in claim 33, wherein said first one reagent tray comprises a plurality of receptacles, each for an aliquot of a substance for reaction with said sample;
a first said receptacle and an second said receptacle define a barrier disposed to prevent said substance from moving from said first said receptacle to said second said receptacle when said reagent tray is disposed in a first position and said aliquot is less than a selected amount, and disposed to not prevent said substance from moving from said first said receptacle to said second said receptacle when said reagent tray is disposed in said first position and said aliquot is greater than said selected amount.
38. A system as in claim 37, wherein said selected amount is just large enough to cover said sample mounted on a slide by capillary action.
39. A system as in claim 33, wherein said first one reagent tray comprises a plurality of receptacles, each for an aliquot of a substance for reaction with said sample;
said sample holder comprises means for holding a plurality of samples; and each said receptacle is disposed to receive exactly one of said plurality of samples coupled to a sample holder when said sample holder is disposed adjacent to said tile.
40. A system as in claim 33, wherein said first one reagent tray comprises a receptacle for an aliquot of a substance for reaction with said sample, said receptacle having a surface/volume ratio low enough to minimize evaporation of said substance.
41. A system as in claim 33, wherein said first one reagent tray comprises a receptacle for an aliquot of a substance for reaction with said sample, said aliquot having a volume just large enough to cover said sample mounted on a slide by capillary action.
42. A system as in claim 33, wherein said first one reagent tray comprises a receptacle for an aliquot of a substance for reaction with said sample, said receptacle having a volume just large enough to receive a slide comprising said sample.
43. A system as in claim 33, wherein said first one reagent tray comprises a rectilinear external form with a reagent tray lock coupled thereto;
said first one reagent tray is disposed for coupling to a first one of said plurality of tiles, said first one tile having a tray receptacle defining a rectilinear region into which said rectilinear external form may just fit;
said tray receptacle comprises a lock receptacle for receiving said reagent tray lock, said lock receptacle defining a void disposed near said rectilinear region into which said reagent tray lock may just fit; and said reagent tray lock is disposed to just fit into said void when said reagent tray is just fit into said tray receptacle.
44. A system as in claim 43, wherein said reagent tray lock comprises a spring member coupled to said rectilinear external form, and a lip member protruding from said rectilinear external form; and said lip member is disposed to just fit into said void when said reagent tray is just fit into said tray receptacle, and said lip member is disposed to prevent release of said reagent tray from said tile in response to a vertical force.
45. A system as in claim 44, wherein said reagent tray comprises a second member protruding from said rectilinear external form;
said tray receptacle comprises a second receptacle for receiving said second member, said second receptacle defining a second void disposed near said rectilinear region into which said second member may just fit;
said second member is disposed to just fit into said second void when said reagent tray is just fit into said tray receptacle; and said reagent tray lock is disposed to allow release of said reagent tray from said tile in response to horizontal force combined with a vertical force.
46. A system as in claim 33, wherein said first one reagent tray is coupled to a first one of said plurality of tiles; and said first one reagent tray comprises a lock coupled to said first one tile.
47. A system as in claim 23, wherein said robotic device comprises a substrate to which are coupled a plurality of workstations, each one of said workstations disposed at a selected location on said substrate and each said workstation having been prefilled with an amount of a substance, said amount and said substance selected from a predetermined set of amounts and substances;
a plurality of analysis programs from a set of predetermined analysis programs stored in a computer memory; and an interleaving program stored in a computer memory that performs said plurality of analysis programs simultaneously by directing a robotic device to move said samples among said plurality of reagent trays, interleaving a plurality of steps of said analysis programs.
48. A system as in claim 23, wherein said robotic device comprises a substrate to which are coupled a plurality of workstations, each one of said workstations disposed at a selected location on said substrate and each said workstation having been prefilled with an amount of a substance, said amount and said substance selected from a predetermined set of amounts and substances;
a robotic arm disposed for reaching substantially all of said tiles and comprising means for coupling to and decoupling from a sample holder, said sample holder having means for holding at least one sample in a selected position, whereby said robotic arm is capable of moving said sample from a first one of said workstations to a second one of said workstations.
49. A system for performing a plurality of independent analysis procedures simultaneously, each said procedure having a sample and at least one process step for operating on that sample, said system comprising means for coupling a plurality of tiles to a substrate, each at a selected location on said substrate;
means for coupling a plurality of sealed reagent trays, each having a substantially uniform shape and size selected from a predetermined set of shapes and sizes, each having been prefilled with an amount of a substance, said amount and said substance selected from a predetermined set of amounts and substances to said plurality of tiles;
means for exposing said substance in said sealed reagent trays while said sealed reagent trays are coupled to said plurality of tiles;
means for selecting a plurality of analysis programs from a set of predetermined analysis programs stored in a computer memory; and means for initiating an interleaving program stored in a computer memory that performs said plurality of analysis programs simultaneously by directing a robotic device to move said samples among said plurality of reagent trays, interleaving a plurality of steps of said analysis programs.
50. In a system for performing a plurality of independent analysis procedures simultaneously, each said procedure having a sample and at least one process step for operating on that sample, a combination of a reagent tray and a tile comprising a reagent tray lock having a spring disposed on said reagent tray and a receiving element for said spring disposed on said tile;
a cover disposed on said reagent tray with adhering means having sufficient power to prevent release of said cover during normal handling and shipping, but able to release said cover when said reagent tray is locked into said tile with said reagent tray lock.
51. A combination as in claim 50, wherein said reagent tray comprises a plurality of receptacles, each for an aliquot of a substance for reaction with a sample;
a first said receptacle and an second said receptacle define a barrier disposed to prevent said substance from moving from said first said receptacle to said second said receptacle when said reagent tray is disposed in a first position, and disposed to not prevent said substance from moving from said first said receptacle to said second said receptacle when said reagent tray is disposed in a second position.
52. A combination as in claim 50, wherein said reagent tray comprises a plurality of receptacles, each for an aliquot of a substance for reaction with a sample;
a first said receptacle and an second said receptacle define a barrier disposed to prevent said substance from moving from said first said receptacle to said second said receptacle when said reagent tray is disposed in a first position and said aliquot is less than a selected amount, and disposed to not prevent said substance from moving from said first said receptacle to said second said receptacle when said reagent tray is disposed in said first position and said aliquot is greater than said selected amount.
53. A combination as in claim 52, wherein said selected amount is just large enough to cover said sample mounted on a slide by capillary action.
54. A combination as in claim 50, wherein said reagent tray comprises a receptacle for an aliquot of a substance for reaction with a sample, said receptacle having a surface/volume ratio low enough to minimize evaporation of said substance.
55. A combination as in claim 50, wherein said reagent tray comprises a receptacle for an aliquot of a substance for reaction with a sample, said aliquot having a volume just large enough to cover said sample mounted on a slide by capillary action.
56. A combination as in claim 50, wherein said reagent tray comprises a receptacle for an aliquot of a substance for reaction with a sample, said receptacle having a volume just large enough to receive a slide comprising said sample.
57. A combination as in claim 50, wherein said reagent tray comprises a rectilinear external form with said reagent tray lock coupled thereto;
said reagent tray is disposed for coupling to a first one of said plurality of tiles, said first one tile having a tray receptacle defining a rectilinear region into which said rectilinear external form may just fit;
said receiving element a lock receptacle for receiving said reagent tray lock, said lock receptacle defining a void disposed near said rectilinear region into which said reagent tray lock may just fit; and said reagent tray lock is disposed to just fit into said void when said reagent tray is just fit into said tray receptacle.
58. A combination as in claim 57, wherein said reagent tray lock comprises a spring member coupled to said rectilinear external form, and a lip member protruding from said rectilinear external form; and said lip member is disposed to just fit into said void when said reagent tray is just fit into said tray receptacle, and said lip member is disposed to prevent release of said reagent tray from said tile in response to a vertical force.
59. A combination as in claim 58, wherein said reagent tray comprises a second member protruding from said rectilinear external form;
said tray receptacle comprises a second receptacle for receiving said second member, said second receptacle defining a second void disposed near said rectilinear region into which said second member may just fit;
said second member is disposed to just fit into said second void when said reagent tray is just fit into said tray receptacle; and said reagent tray lock is disposed to allow release of said reagent tray from said tile in response to horizontal force combined with a vertical force.
60. In a system for performing a plurality of independent analysis procedures simultaneously each said procedure having a sample and at least one process step for operating on that sample, a data structure comprising a sequence of process steps indexed by a time value and indicating a start time and an end time for each said process step.
61. A data structure as in claim 60, comprising a second sequence of process steps indexed by said time value, wherein said second sequence comprises process steps from a single procedure.
62. In a system for performing a plurality of independent analysis procedures simultaneously, each said procedure having a sample and at least one process step for operating on that sample, a data structure comprising an ordered sequence of entries, each said entry comprising a symbol on a display screen and a process step.
63. A data structure as in claim 62, wherein each entry comprises timing information for said process step.
64. In a system for performing a plurality of independent analysis procedures simultaneously, each said procedure having a sample and at least one process step for operating on that sample, a device for coupling a robotic hand to a slide holder, comprising a first horizontal element defining a first hole adapted to be coupled to said robotic hand;
a second horizontal element disposed above said first horizontal element and adapted to restrain said first horizontal element from motion in a vertical direction, and defining a second hole;
a third horizontal element disposed below said first horizontal element and adapted to restrain said first horizontal element from motion in a vertical direction, and defining a third hole;
said second hole disposed with at least some exposed area that is not exposed by said first hole, and said third hole disposed with at least some exposed are that is not exposed by said first hole, whereby said first horizontal element is free to move in response to coupling by said robotic hand.
65. A device as in claim 64, comprising a bumper disposed above said first horizontal element.
66. A device as in claim 64, comprising a guiding member coupled to said first horizontal element, whereby small misalignments by said robotic hand cause said first horizontal element to move in response to coupling by said robotic hand.
67. A device as in claim 66, wherein said guiding member comprises a lip surrounding at least a part of said first hole.
68. A device as in claim 64, comprising means for coupling said second horizontal element to a frame.
69. A device as in claim 64, comprising means for coupling said second horizontal element to said third horizontal element, whereby said second horizontal element and said third horizontal element define a space for disposing said first horizontal element.
70. A device as in claim 64, wherein at least one of said second and third holes is substantially elliptical.
71. A device as in claim 64, wherein said first hole is substantially elliptical.
72. A device as in claim 64, wherein said first horizontal element comprises a disk.
73. A device as in claim 64, wherein said first horizontal element is substantially thinner than at least one of said second horizontal element and said third horizontal element.
74. In a system for performing a plurality of independent analysis procedures simultaneously, each said procedure having a sample and at least one process step for operating on that sample, a hydration fluid comprising less than about 3% of a surfactant.
75. A hydration fluid as in claim 74, comprising about .2% of said surfactant and about .1% of an acid.
76. A hydration fluid as in claim 74, comprising about 9980 milliliters water, about 20 milliliters tween, and about 10 grams sorbic acid, per 10 liters of fluid.
77. A hydration fluid as in claim 74, comprising less than about 3% of an acid.
78. A hydration fluid as in claim 74, wherein said surfactant is bridge or tween.
79. In a system for performing a plurality of independent analysis procedures simultaneously, each said procedure having a sample and at least one process step for operating on that sample, an incubation oven element comprising a hydration fluid supply;
an incubation chamber coupled to said hydration fluid supply, said incubation chamber comprising a heat exchanger;
means for regulating an amount of said hydration fluid applied to said sample; and means for regulating a temperature applied to said sample by said heat exchanger.
80. In a system for performing a plurality of independent analysis procedures simultaneously, each said procedure having a sample and at least one process step for operating on that sample, a reagent tray comprising a rectilinear external form with a reagent tray lock coupled thereto;
said reagent tray lock disposed for coupling to a tile, said tile being disposed at a selected location on a substrate in said system;
said tile having a tray receptacle defining a rectilinear region into which said rectilinear external form may just fit;
said tray receptacle comprises a lock receptacle for receiving said reagent tray lock, said lock receptacle defining a void disposed near said rectilinear region into which said reagent tray lock may just fit; and said reagent tray lock is disposed to just fit into said void when said reagent tray is just fit into said tray receptacle.
81. A reagent tray as in claim 80, wherein said reagent tray comprises a cover disposed thereon with adhering means having sufficient power to prevent release of said cover during normal handling and shipping, but able to release said cover when said reagent tray is locked into said tile with said reagent tray lock.
82. A reagent tray as in claim 80, wherein said reagent tray comprises a plurality of receptacles, each for an aliquot of a substance for reaction with said sample;

a first said receptacle and an second said receptacle define a barrier disposed to prevent said substance from moving from said first said receptacle to said second said receptacle when said reagent tray is disposed in a first position, and disposed to not prevent said substance from moving from said first said receptacle to said second said receptacle when said reagent tray is disposed in a second position.
83. A reagent tray as in claim 80, wherein said reagent tray comprises a plurality of receptacles, each for an aliquot of a substance for reaction with said sample;
a first said receptacle and an second said receptacle define a barrier disposed to prevent said substance from moving from said first said receptacle to said second said receptacle when said reagent tray is disposed in a first position and said aliquot is less than a selected amount, and disposed to not prevent said substance from moving from said first said receptacle to said second said receptacle when said reagent tray is disposed in said first position and said aliquot is greater than said selected amount.
84. A reagent tray as in claim 83, wherein said selected amount is just large enough to cover said sample mounted on a slide by capillary action.
85. A reagent tray as in claim 80, wherein said reagent tray comprises a plurality of receptacles, each for an aliquot of a substance for reaction with said sample;

said system comprises a sample holder having means for holding a plurality of samples; and each said receptacle is disposed to receive exactly one of said plurality of samples coupled to a sample holder when said sample holder is disposed adjacent to said tile.
86. A reagent tray as in claim 80, wherein said reagent tray comprises a receptacle for an aliquot of a substance for reaction with said sample, said receptacle having a surface/volume ratio low enough to minimize evaporation of said substance.
87. A reagent tray as in claim 80, wherein said reagent tray comprises a receptacle for an aliquot of a substance for reaction with said sample, said aliquot having a volume just large enough to cover said sample mounted on a slide by capillary action.
88. A reagent tray as in claim 80, wherein said reagent tray comprises a receptacle for an aliquot of a substance for reaction with said sample, said receptacle having a volume just large enough to receive a slide comprising said sample.
89. A reagent tray as in claim 80, wherein said reagent tray lock comprises a spring member coupled to said rectilinear external form, and a lip member protruding from said rectilinear external form; and said lip member is disposed to just fit into said void when said reagent tray is just fit into said tray receptacle, and said lip member is disposed to prevent release of said reagent tray from said tile in response to a vertical force.
90. A reagent tray as in claim 89, wherein said reagent tray comprises a second member protruding from said rectilinear external form;
said tray receptacle comprises a second receptacle for receiving said second member, said second receptacle defining a second void disposed near said rectilinear region into which said second member may just fit;
said second member is disposed to just fit into said second void when said reagent tray is just fit into said tray receptacle; and said reagent tray lock is disposed to allow release of said reagent tray from said tile in response to horizontal force combined with a vertical force.
91. In a system for performing a plurality of independent analysis procedures simultaneously, each said procedure having a sample and at least one process step for operating on that sample, a workstation comprising an incubation oven element comprising a hydration fluid supply, an incubation chamber coupled to said hydration fluid supply, said incubation chamber comprising a heat exchanger having a plurality of heat exchanger elements disposed in an array, means for regulating an amount of said hydration fluid applied to said sample, and means for regulating a temperature applied to said sample by said heat exchanger; and a slide holder adapted for disposition at said incubation oven element, said slide holder comprising a set of slides disposed in an array matched to said heat exchanger element.
92. A workstation as in claim 91, comprising means for cooling said incubation chamber while said slides are disposed therein.
93. A workstation as in claim 92, wherein said means for cooling comprises a fan and a temperature regulator coupled thereto.
94. A workstation as in claim 91, comprising means for isolating said heat exchanger elements from said hydration fluid.
95. A workstation as in claim 91, comprising one said heat exchanger element for each pair of said slides.
96. A workstation as in claim 91, wherein said heat exchanger elements comprise fins adapted to be disposed near to said samples on said slides.
97. A workstation as in claim 91, wherein said means for regulating an amount of said hydration fluid comprises a fluid well coupled to said incubation chamber; and means for maintaining a level of hydration fluid in said fluid well at a selected level.
98. A workstation as in claim 91, wherein said means for regulating a temperature comprises means for cooling said incubation chamber.
99. A workstation as in claim 91, wherein said slide holder comprises a slide cover having a set of adjustable ventilation openings.
100. A workstation as in claim 91, wherein said slide holder comprises a V shaped structural member disposed to direct condensation away from a set of slides.
CA002191776A 1994-05-31 1995-05-17 Automated tissue assay using standardized chemicals and packages Abandoned CA2191776A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US08/252,282 US5696887A (en) 1991-08-05 1994-05-31 Automated tissue assay using standardized chemicals and packages
US08/252,282 1994-05-31

Publications (1)

Publication Number Publication Date
CA2191776A1 true CA2191776A1 (en) 1995-12-07

Family

ID=22955357

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002191776A Abandoned CA2191776A1 (en) 1994-05-31 1995-05-17 Automated tissue assay using standardized chemicals and packages

Country Status (6)

Country Link
US (5) US5696887A (en)
EP (1) EP0804730A1 (en)
JP (1) JPH10504892A (en)
AU (1) AU2638995A (en)
CA (1) CA2191776A1 (en)
WO (1) WO1995033240A1 (en)

Families Citing this family (85)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5696887A (en) * 1991-08-05 1997-12-09 Biotek Solutions, Incorporated Automated tissue assay using standardized chemicals and packages
AU4318697A (en) * 1996-09-18 1998-04-14 Kabushiki Kaisha Tiyoda Seisakusho Liquid treating apparatus for biological sample
US5839091A (en) * 1996-10-07 1998-11-17 Lab Vision Corporation Method and apparatus for automatic tissue staining
US6735531B2 (en) 1996-10-07 2004-05-11 Lab Vision Corporation Method and apparatus for automatic tissue staining
DE19652339A1 (en) * 1996-12-17 1998-06-18 Microm Laborgeraete Gmbh Device for treating objects
US5985214A (en) 1997-05-16 1999-11-16 Aurora Biosciences Corporation Systems and methods for rapidly identifying useful chemicals in liquid samples
US6207408B1 (en) * 1997-08-20 2001-03-27 University Of Miami High quality, continuous throughput, tissue fixation-dehydration-fat removal-impregnation method
US6793890B2 (en) * 1997-08-20 2004-09-21 The University Of Miami Rapid tissue processor
EP0962775A1 (en) * 1998-04-28 1999-12-08 Nevio Recinelli Automatic analyzer with single robot arm and U-shaped reaction capillary vessels
CA2273729A1 (en) 1998-07-14 2000-01-14 Bayer Corporation Robotics for transporting containers and objects within an automated analytical instrument and service tool for servicing robotics
US8298485B2 (en) * 1999-07-08 2012-10-30 Lee H. Angros In situ heat induced antigen recovery and staining apparatus and method
US7951612B2 (en) * 1999-07-08 2011-05-31 Lee H. Angros In situ heat induced antigen recovery and staining apparatus and method
WO2001004634A1 (en) * 1999-07-08 2001-01-18 Lee Angros Antigen recovery and/or staining apparatus and method
US7897106B2 (en) * 1999-07-08 2011-03-01 Lee Angros Situ heat induced antigen recovery and staining apparatus and method
KR100675698B1 (en) 1999-08-06 2007-02-01 써모 바이오스타, 인크. An automated point of care detection system including complete sample processing capabilities
WO2001044784A1 (en) * 1999-12-14 2001-06-21 University Of Miami Microwave unit and system for tissue processing
DE10041226A1 (en) * 2000-08-22 2002-03-07 Leica Microsystems Method for treatment of cytological and histological specimens, selects shortest paths for distributing specimens to group of treatment stations
DE10052833A1 (en) * 2000-10-24 2002-04-25 Leica Microsystems Laboratory assembly to dye cytological or histological preparations monitors reagents over extended period
DE10052832A1 (en) * 2000-10-24 2002-04-25 Leica Microsystems Laboratory assembly treating cytological or histological preparations has series of identical treatment stations for use as required
DE10052834B4 (en) * 2000-10-24 2019-09-19 Leica Biosystems Nussloch Gmbh Method of treating objects
EP1243911A1 (en) * 2001-03-22 2002-09-25 Leica Mikrosysteme GmbH Device for tissue preparation
EP1248170B1 (en) * 2001-04-05 2009-09-16 INPECO IP Ltd. Method for the management of workcell systems based on an automation management system
US7270785B1 (en) * 2001-11-02 2007-09-18 Ventana Medical Systems, Inc. Automated molecular pathology apparatus having fixed slide platforms
WO2003042788A2 (en) 2001-11-13 2003-05-22 Chromavision Medical Systems, Inc. A system for tracking biological samples
US6998270B2 (en) * 2001-11-26 2006-02-14 Lab Vision Corporation Automated tissue staining system and reagent container
US20040033163A1 (en) * 2001-11-26 2004-02-19 Lab Vision Corporation Automated tissue staining system and reagent container
US20030186353A1 (en) * 2002-04-02 2003-10-02 Page Erickson Microscopic precision construction of tissue array block
EP1494808B1 (en) * 2002-04-15 2013-07-03 Ventana Medical Systems, Inc. Automated high volume slide staining system
US11249095B2 (en) 2002-04-15 2022-02-15 Ventana Medical Systems, Inc. Automated high volume slide processing system
US7468161B2 (en) 2002-04-15 2008-12-23 Ventana Medical Systems, Inc. Automated high volume slide processing system
WO2003091710A1 (en) * 2002-04-26 2003-11-06 Ventana Medical Systems, Inc. Automated molecular pathology apparatus having fixed slide platforms
JP2005533255A (en) * 2002-07-16 2005-11-04 ベンタナ・メデイカル・システムズ・インコーポレーテツド Automated histochemical processing tray
US7648678B2 (en) 2002-12-20 2010-01-19 Dako Denmark A/S Method and system for pretreatment of tissue slides
US7584019B2 (en) * 2003-12-15 2009-09-01 Dako Denmark A/S Systems and methods for the automated pre-treatment and processing of biological samples
US7850912B2 (en) * 2003-05-14 2010-12-14 Dako Denmark A/S Method and apparatus for automated pre-treatment and processing of biological samples
US7875245B2 (en) * 2003-05-14 2011-01-25 Dako Denmark A/S Method and apparatus for automated pre-treatment and processing of biological samples
AU2003900810A0 (en) * 2003-02-24 2003-03-13 Vision Biosystems Limited Method of scheduling
US20050074890A1 (en) * 2003-05-19 2005-04-07 Ventana Medical Systems, Inc. Electrophoretic in situ tissue staining
US7642064B2 (en) 2003-06-24 2010-01-05 Ventana Medical Systems, Inc. Enzyme-catalyzed metal deposition for the enhanced detection of analytes of interest
JP4648902B2 (en) * 2003-06-24 2011-03-09 ベンタナ・メデイカル・システムズ・インコーポレーテツド Enzyme-catalyzed metal attachment for improved in situ detection of immunohistochemical epitopes and nucleic acid sequences
US20080235055A1 (en) * 2003-07-17 2008-09-25 Scott Mattingly Laboratory instrumentation information management and control network
US8719053B2 (en) * 2003-07-17 2014-05-06 Ventana Medical Systems, Inc. Laboratory instrumentation information management and control network
US7860727B2 (en) 2003-07-17 2010-12-28 Ventana Medical Systems, Inc. Laboratory instrumentation information management and control network
US7767152B2 (en) 2003-08-11 2010-08-03 Sakura Finetek U.S.A., Inc. Reagent container and slide reaction retaining tray, and method of operation
US7744817B2 (en) 2003-08-11 2010-06-29 Sakura Finetek U.S.A., Inc. Manifold assembly
US9518899B2 (en) 2003-08-11 2016-12-13 Sakura Finetek U.S.A., Inc. Automated reagent dispensing system and method of operation
US7501283B2 (en) * 2003-08-11 2009-03-10 Sakura Finetek U.S.A., Inc. Fluid dispensing apparatus
DE10344284A1 (en) * 2003-09-24 2005-05-04 Keyneurotek Ag Device and method for the automated performance of laboratory work steps
US7470401B2 (en) * 2003-10-24 2008-12-30 The University Of Miami Simplified tissue processing
US8026106B1 (en) 2003-12-18 2011-09-27 Ventana Medical Systems, Inc. Removal of mercuric chloride crystals from fixed tissues
EP1697719B1 (en) * 2003-12-23 2018-09-19 Ventana Medical Systems, Inc. Method and apparatus for efficient thin film fluid processing of flat surfaces
CA2556772A1 (en) 2004-03-02 2005-09-15 Dako Denmark A/S Reagent delivery system, dispensing device and container for a biological staining apparatus
AU2005259981B2 (en) 2004-06-29 2012-01-12 Dako Denmark A/S Method of pre-treatment and staining of and support device for a biological sample
US7867443B2 (en) * 2004-07-23 2011-01-11 Dako Denmark A/S Method and apparatus for automated pre-treatment and processing of biological samples
EP1771730B1 (en) * 2004-07-23 2014-08-20 Ventana Medical Systems, Inc. Method and apparatus for applying fluids to a biological sample
GB0501590D0 (en) * 2005-01-25 2005-03-02 Ceres Power Ltd Processing of enhanced performance LSCF fuel cell cathode microstructure and a fuel cell cathode
US7680559B2 (en) * 2005-02-08 2010-03-16 Lam Research Corporation Wafer movement control macros
CA2623241A1 (en) * 2005-04-21 2006-11-02 Celerus Diagnostics, Inc. Parallel processing fluidic method and apparatus for automated rapid immunohistochemistry
WO2006127852A2 (en) 2005-05-24 2006-11-30 Angros Lee H In situ heat induced antigen recovery and staining apparatus and method
EP1904670B1 (en) 2005-06-28 2011-09-28 The Board of Regents of the University of Oklahoma Methods for growing and harvesting carbon nanotubes
KR100749579B1 (en) * 2005-09-05 2007-08-16 삼성광주전자 주식회사 Moving Robot having a plurality of changeable work module and Control Method for the same
US7853356B2 (en) * 2006-04-14 2010-12-14 Fanuc Robotics America, Inc. Method for optimizing a robot program and a robot system
US8459509B2 (en) 2006-05-25 2013-06-11 Sakura Finetek U.S.A., Inc. Fluid dispensing apparatus
US7875242B2 (en) * 2006-10-17 2011-01-25 Preyas Sarabhai Shah Slide stainer with multiple heater stations
DE102007008713B4 (en) 2007-02-20 2019-07-11 Leica Biosystems Nussloch Gmbh Tissue infiltration device
WO2009106081A1 (en) 2008-02-29 2009-09-03 Dako Denmark A/S Systems and methods for tracking and providing workflow information
US20090298172A1 (en) 2008-05-28 2009-12-03 Steven Paul Wheeler Histological specimen treatment apparatus and method
EP2148206B1 (en) * 2008-07-25 2015-11-18 F.Hoffmann-La Roche Ag A laboratory system for handling sample tube racks, an alignmemt element for sample tube racks and a rack tray receiver assembly
GB2477648B (en) 2008-08-29 2013-02-20 Lee H Angros In situ heat induced antigen recovery and staining apparatus and method
AU2009313985B2 (en) 2008-11-12 2014-07-31 Ventana Medical Systems, Inc. Methods and apparatuses for heating slides carrying specimens
KR101812820B1 (en) * 2010-01-08 2017-12-27 웨이크 포리스트 유니버시티 헬스 사이언시즈 Delivery system
JP5432816B2 (en) * 2010-05-14 2014-03-05 株式会社日立ハイテクノロジーズ Automatic analysis system and device management server
JP5591742B2 (en) * 2010-06-30 2014-09-17 シスメックス株式会社 Specimen Testing Device Processing Capacity Information Generation Device, Specimen Testing Apparatus, Specimen Testing Device Processing Capacity Information Generation Method, and Computer Program
US8752732B2 (en) 2011-02-01 2014-06-17 Sakura Finetek U.S.A., Inc. Fluid dispensing system
WO2012122105A1 (en) 2011-03-07 2012-09-13 Wake Forest University Health Sciences Delivery system
US8932543B2 (en) 2011-09-21 2015-01-13 Sakura Finetek U.S.A., Inc. Automated staining system and reaction chamber
US8580568B2 (en) 2011-09-21 2013-11-12 Sakura Finetek U.S.A., Inc. Traceability for automated staining system
US9091621B2 (en) 2013-04-05 2015-07-28 GGB Company Multiple slide processing apparatus
AU2014363717B2 (en) 2013-12-13 2016-12-22 Ventana Medical Systems, Inc. Automated histological processing of biological specimens and associated technology
JP6432883B2 (en) * 2014-11-21 2018-12-05 国立研究開発法人産業技術総合研究所 Operation command generation device, operation command generation method, computer program, and processing system
JP6419547B2 (en) 2014-11-21 2018-11-07 国立研究開発法人産業技術総合研究所 Operation command generation device, operation command generation method, computer program, and processing system.
US10365189B2 (en) 2015-05-07 2019-07-30 Steven Wheeler Histological specimen treatment
EP4053565A1 (en) * 2015-09-11 2022-09-07 Hitachi High-Tech Corporation Automated analyzer
US10768190B2 (en) 2017-01-27 2020-09-08 Lonza Limited Dynamically controlling an automated system
CN109253874B (en) * 2018-10-19 2020-03-31 日照职业技术学院 Robot arm flexibility ratio detection device

Family Cites Families (80)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3665148A (en) * 1971-04-07 1972-05-23 Gen Motors Corp Six-axis manipulator
US4058367A (en) 1976-05-19 1977-11-15 Gilford Instrument Laboratories Inc. Automatic asynchronous fluid processing apparatus
SU603577A1 (en) * 1976-07-06 1978-04-25 Ордена Ленина, Ордена Октябрьской Революции И Ордена Трудового Красного Знамени Завод "Ростсельмаш" Industrial robot
US4087248A (en) * 1976-07-26 1978-05-02 Miles Laughton E Multiple assay machine and method
JPS5514157A (en) * 1978-07-14 1980-01-31 Shin Meiwa Ind Co Ltd Automatic welder
JPS55136958A (en) 1979-04-14 1980-10-25 Olympus Optical Co Ltd Automatic analyzer
US5037484A (en) * 1981-04-20 1991-08-06 Alcon Laboratories, Inc. Cleaning agent for optical surfaces
IT1144709B (en) * 1981-05-15 1986-10-29 Dea Spa DIMENSIONAL MEASUREMENT SYSTEM SERVED BY A MULTIPLE OF OPERATING ARMS AND CONTROLLED BY A CALCULATOR SYSTEM
US4384193A (en) * 1981-06-09 1983-05-17 Immulok, Inc. Incubating device for specimen mounted on glass slides in immunoassays
JPS5848836A (en) * 1981-09-18 1983-03-22 Toa Medical Electronics Co Ltd Optical type automatic analyzing and measuring device
US4727409A (en) 1982-04-12 1988-02-23 Signetics Corporation Programmable read-only memory formed with opposing PN diodes
DE3473512D1 (en) * 1983-04-15 1988-09-22 Agency Science & Tech Chemical manipulator
GB2143205B (en) * 1983-07-15 1986-07-16 Leicester Polytechnic Robots
JPS6111293A (en) 1984-06-27 1986-01-18 Kanzaki Paper Mfg Co Ltd Image receiving sheet for thermal transfer recording
JPS6149205A (en) * 1984-08-16 1986-03-11 Seiko Instr & Electronics Ltd Robot control system
JPH0690211B2 (en) 1984-09-21 1994-11-14 オリンパス光学工業株式会社 Immunological analyzer and method thereof
US4952518A (en) * 1984-10-01 1990-08-28 Cetus Corporation Automated assay machine and assay tray
US4727494A (en) * 1985-01-01 1988-02-23 Zymark Corporation Computerized robot control system with scheduling feature
US4648023A (en) 1985-05-23 1987-03-03 Powell Roger A Method for resource allocation for the manufacture of a product
US4719087A (en) * 1985-07-01 1988-01-12 Baxter Travenol Laboratories, Inc. Tray for analyzing system
US5023187A (en) 1985-09-13 1991-06-11 Fisher Scientific Company Method and device for accelerated treatment of thin sample on surface
US4731335A (en) 1985-09-13 1988-03-15 Fisher Scientific Company Method for treating thin samples on a surface employing capillary flow
US4670974A (en) 1985-11-06 1987-06-09 Westinghouse Electric Corp. Windshield insertion system for a vehicle on a moving conveyor apparatus
US4678752A (en) 1985-11-18 1987-07-07 Becton, Dickinson And Company Automatic random access analyzer
GB8605324D0 (en) * 1986-03-04 1986-04-09 Rank Taylor Hobson Ltd Metrological apparatus
US4843566A (en) * 1986-03-07 1989-06-27 Hewlett-Packard Company Robot motion control system
WO1987006008A2 (en) * 1986-03-26 1987-10-08 Beckman Instruments, Inc. Automated multi-purposse analytical chemistry processing center and laboratory work station
US4835711A (en) * 1986-05-30 1989-05-30 Zymark Corporation Quickly reconfigurable robotic system
FR2600166B1 (en) 1986-06-17 1988-10-07 Rhone Poulenc Rech PROCESS AND DEVICE FOR AUTOMATICALLY SUPPORTING AND ANALYZING SAMPLES OF PRODUCTS RANDOMIZED
EP0255026A3 (en) * 1986-07-23 1988-10-19 Takeda Chemical Industries, Ltd. Automatic analysis method and apparatus for enzyme reaction
JP2533495B2 (en) * 1986-07-25 1996-09-11 株式会社日立製作所 Work scheduling method and apparatus
FR2605105B1 (en) * 1986-10-14 1989-08-04 Tiyoda Seisakusho Kk SAMPLE COLORING APPARATUS FOR MICROSCOPE EXAMINATION
GR871619B (en) 1986-10-31 1988-03-03 Genetic Systems Corp Automated patient sample analysis instrument
EP0275826A1 (en) * 1986-12-16 1988-07-27 Ciba-Geigy Ag Control system for a sample preparation system
JPS63208761A (en) 1987-02-25 1988-08-30 Chiyoda Seisakusho:Kk Method and device for obtaining procedure to execute plural kinds of staining methods simultaneously by automatic staining device for microscopic specimen
US4954237A (en) * 1987-03-16 1990-09-04 Helena Laboratories Automatic electrophoresis apparatus
US5002736A (en) 1987-03-31 1991-03-26 Fisher Scientific Co. Microscope slide and slide assembly
US4803048A (en) * 1987-04-02 1989-02-07 Nason Frederic L Laboratory kit
US5148370A (en) * 1987-06-17 1992-09-15 The Standard Oil Company Expert system and method for batch production scheduling and planning
JPS645779A (en) * 1987-06-29 1989-01-10 Fanuc Ltd Robot arrangement examination system
US4869813A (en) * 1987-07-02 1989-09-26 Northrop Corporation Drill inspection and sorting method and apparatus
US4979093A (en) * 1987-07-16 1990-12-18 Cavro Scientific Instruments XYZ positioner
US4896269A (en) * 1988-02-29 1990-01-23 General Electric Company Job shop scheduling and production method and apparatus
FR2630216A1 (en) * 1988-04-14 1989-10-20 Ibal Robotised installation for analyses, in particular medical analyses
US4929619A (en) * 1988-07-14 1990-05-29 Thames Pharmacal Co., Inc. Anti-infective methods and compositions
US5229074A (en) * 1988-07-25 1993-07-20 Precision Systems, Inc. Automatic multiple-sample multiple-reagent chemical analyzer
EP0353589B1 (en) 1988-08-02 1996-02-07 Abbott Laboratories Apparatus and method for providing assay calibration data
US5016170A (en) * 1988-09-22 1991-05-14 Pollalis Spiro N Task management
US5122959A (en) 1988-10-28 1992-06-16 Automated Dispatch Services, Inc. Transportation dispatch and delivery tracking system
DE68910304T2 (en) * 1989-03-15 1994-02-24 Hewlett Packard Co Follow-up rules for pre-injecting samples to be analyzed by chromatography.
JPH02284885A (en) * 1989-04-27 1990-11-22 Nissan Motor Co Ltd Position teaching method for workpiece positioning device
US5281394A (en) * 1989-07-21 1994-01-25 Icn Biomedicals, Inc. Data collection and sample handling apparatus
JPH0363568A (en) 1989-07-31 1991-03-19 Mitsui Toatsu Chem Inc Monitoring system for automatic analysis of liquid resin
US4975250A (en) 1989-08-21 1990-12-04 Fisher Scientific Co. Aligned slideholder and assembly
US5040123A (en) 1989-09-08 1991-08-13 General Motors Corporation Expert system scheduler
DE69030318T2 (en) * 1989-10-17 1997-10-16 Perkin Elmer Corp ROBOTIC INTERFACE
US5233533A (en) 1989-12-19 1993-08-03 Symmetrix, Inc. Scheduling method and apparatus
US5355304A (en) * 1990-01-30 1994-10-11 Demoranville Victoria E Clinical laboratory work-flow system which semi-automates validated immunoassay and electrophoresis protocols
US5595707A (en) * 1990-03-02 1997-01-21 Ventana Medical Systems, Inc. Automated biological reaction apparatus
US5225325A (en) * 1990-03-02 1993-07-06 Ventana Medical Systems, Inc. Immunohistochemical staining method and reagents therefor
US5168441A (en) * 1990-05-30 1992-12-01 Allen-Bradley Company, Inc. Methods for set up and programming of machine and process controllers
US5272093A (en) * 1990-08-02 1993-12-21 Miles Inc. Reagent containment and delivery tray and method of use
US5282149A (en) * 1991-06-03 1994-01-25 Abbott Laboratories Adaptive scheduling system and method for a biological analyzer with reproducible operation time periods
US5576215A (en) 1991-06-03 1996-11-19 Abbott Laboratories Adaptive scheduling system and method for operating a biological sample analyzer with variable interval periods
US5246665A (en) 1991-06-03 1993-09-21 Abbott Laboratories Heat and air flow control for assay carrier
US5402350A (en) * 1991-06-28 1995-03-28 Texas Instruments Incorporated Scheduling for multi-task manufacturing equipment
US5696887A (en) * 1991-08-05 1997-12-09 Biotek Solutions, Incorporated Automated tissue assay using standardized chemicals and packages
US5355439A (en) * 1991-08-05 1994-10-11 Bio Tek Instruments Method and apparatus for automated tissue assay
EP0531811B1 (en) * 1991-08-30 1997-04-23 Canon Kabushiki Kaisha Hand mechanism for robot
IT1260447B (en) * 1992-01-27 1996-04-09 PROCEDURE AND EQUIPMENT FOR THE INTEGRAL AUTOMATION OF AN ANALYSIS LABORATORY
US5376313A (en) * 1992-03-27 1994-12-27 Abbott Laboratories Injection molding a plastic assay cuvette having low birefringence
US5646049A (en) * 1992-03-27 1997-07-08 Abbott Laboratories Scheduling operation of an automated analytical system
US5380487A (en) * 1992-05-05 1995-01-10 Pasteur Sanofi Diagnostics Device for automatic chemical analysis
US5352612A (en) * 1993-02-09 1994-10-04 Baxter Diagnostics Inc. Method and apparatus for the stepwise movement of items
US5479581A (en) * 1993-12-30 1995-12-26 Storage Technology Corporation Multiple library media exchange system and method
US5578270A (en) * 1995-03-24 1996-11-26 Becton Dickinson And Company System for nucleic acid based diagnostic assay
US5737498A (en) * 1995-07-11 1998-04-07 Beckman Instruments, Inc. Process automation method and apparatus
US5975740A (en) * 1996-05-28 1999-11-02 Applied Materials, Inc. Apparatus, method and medium for enhancing the throughput of a wafer processing facility using a multi-slot cool down chamber and a priority transfer scheme
US5909674A (en) * 1996-12-23 1999-06-01 Philips Electronics North America Corp. Method for optimizing the layout and charge maps of a flowline of pick and place machines
US6068393A (en) * 1997-11-05 2000-05-30 Zymark Corporation Robotic system for processing chemical products

Also Published As

Publication number Publication date
US5737499A (en) 1998-04-07
JPH10504892A (en) 1998-05-12
US5758033A (en) 1998-05-26
WO1995033240A1 (en) 1995-12-07
EP0804730A1 (en) 1997-11-05
US5875286A (en) 1999-02-23
US5696887A (en) 1997-12-09
AU2638995A (en) 1995-12-21
EP0804730A4 (en) 1997-11-05
US6594537B1 (en) 2003-07-15

Similar Documents

Publication Publication Date Title
CA2191776A1 (en) Automated tissue assay using standardized chemicals and packages
EP0600939B1 (en) Method and apparatus for automated tissue assay
US5930461A (en) Method and apparatus for automated tissue assay
EP1247084B1 (en) Method and apparatus for automatic tissue staining
AU2003301103B2 (en) Advanced programmed sample processing system and methods of biological slide processing
EP1446649B1 (en) Method and apparatus for automatic tissue staining
JP5658226B2 (en) Automatic slide processing system
EP1514089B1 (en) Biological reaction apparatus with draining mechanism
US7142987B2 (en) Apparatus, system, and method of archival and retrieval of samples
US6349264B1 (en) Method and apparatus for automatic tissue staining
EP2780688B1 (en) Biological sample treatment apparatus
WO2003040991A2 (en) Archive and analysis system and method
EP1955041A1 (en) Removal of embedding medium
EP1461158A2 (en) Sample carrier comprising a structural array for archiving sample material
EP3910320A1 (en) Computer-implemented method and system for spectroscopic analysis of biological material
Okumus et al. Microfluidics-Assisted Cell Screening (MACS): An automated platform for single-cell microscopy of suspension cultures
CN117558380A (en) High-flux preparation method and system of magnetic micro-nano material based on artificial intelligence algorithm

Legal Events

Date Code Title Description
FZDE Discontinued