What we do

The Immune Analysis Facility (IAF) at Roswell Park Comprehensive Cancer Center is responsible for serial monitoring of immunologic functions in patients with cancer, patients treated with biologic therapies, and patients participating in clinical trials or research protocols at Roswell Park and external institutions and hospitals.

The development of immune monitoring assays is essential to determine the immune responses in patients receiving novel immunotherapies and ultimately transitioning these therapies from the clinical trial phase to standard of care. The IAF offers a broad range of state-of-the-art immunologic assays utilizing multiple platforms to support novel investigator-initiated immunotherapy trials.

Offering the latest immunomonitoring technology

We are committed to continuously expanding access to unique technologies and the use of advanced instrumentation and assisting with experimental design and analysis. As advances in immunobiology occur we will perform pre-clinical evaluations of new assays requested and add them to the available assay list if deemed reliable and standardized.

The IAF offers consultation regarding optimal immunologic assessment and assay development for innovative approaches to evaluate immune responses. We provide expert advice regarding the types of assays for immunomonitoring and data interpretation tailored to fit the endpoints of each specific clinical trial.

Request a service

Interested investigators are asked to provide a summary of their clinical trial design, which will be reviewed by IAF in order to design the appropriate methodology to successfully measure the endpoints of the study.

Services & fees

The Immune Analysis Facility offers a full complement of services designed to monitor patients’ immunological function during cancer treatment to support clinical and translational studies:

Submit a service request

This assay is used to measure the antibody titer specific to cancer antigens. The serially diluted serum or plasma from cancer patients are added in an ELISA plate coated with recombinant protein antigens.

The antibody bound to proteins is detected by utilizing enzyme-labeled anti-human IgG-specific antibody and fluorescent substrate.

In addition to measurement of autoantibodies, commercially available kits can be used for the detection of cytokine and chemokine levels in serum, plasma, fluid or tissue culture supernatants.

The Ella system is a next generation hands-free and high-performance immunoassays, allowing researchers to acquire highly reproducible validated assay data with very few manual steps, which is the only self-contained testing platform for the simultaneous execution of multiple ELISA-based immunoassays.

The assay volume required is 25 μl or less for up to 4-plex customized assays and the data is ready in two hours from sample prep.

This assay is used to detect and quantify the number of T cells that secrete a particular cytokine (e.g. Interferon-γ) upon recognition of a specific antigen.

T cells are cultured with antigen-presenting cells in wells coated with an antibody recognizing a particular cytokine. The coating antibody captures the secreted cytokine and a second cytokine-specific antibody is coupled to a chromogenic substrate and used for detection. Results appear as spots, with each spot corresponding to one cytokine-secreting cell. The number of spots equals the number of cytokine-secreting cells for a specific antigen.

This assay does not determine the amount of cytokine secreted. This assay is highly sensitive (1 cell in 100,000) and can be performed directly ex-vivo using relatively few T cells.

Seahorse XFe96 extracellular flux analyzer tracks and records the oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) that reflects mitochondrial respiration and glycolysis in real-time in 96-well plate format.

Validated assays include the glycolysis stress test, cell mito stress test, mito fuel flex test, glycolytic rate assay, ATP rate assay, cell energy phenotype test and fatty acid oxidation measurement. Custom-made assays are also available.

Helios/CyTOF, a time-of-flight mass cytometry, is capable of acquiring single cell suspension labeled with transition metal isotopes-conjugated monoclonal antibodies.

The main advantages of mass cytometry over conventional fluorescence-based flow cytometry are minimal spectral overlap of each parameter, thereby, the CyTOF can theoretically detect up to 135 isotopes without compensation.

In addition, since the metal isotopes used for conjugation are rare in biological samples and the environment, it can detect the specific signal of each protein on individual cells regardless of autofluorescence.

Currently this system allows single cell analysis with up to 42 parameters for phenotype and function.

Mass Cytometry figure
Mass Cytometry figure

Hyperion Image Mass Cytometer (IMC) is a high-resolution laser ablation system for 1 m cellular resolution, allowing the most comprehensive analysis of the immune and non-immune contexture in the tumor microenvironment.

The IMC module uses the CyTOF instrument to detect up to 40 markers simultaneously on a tissue slide and far exceeds the capabilities of any multi-spectral IHC system that currently exists, by measuring the abundance of metal isotopes tagged to antibodies.

More than 100 metal isotopes tagged antibodies for the IMC are now commercially available for immunocytometry and immunohistochemistry analyses. Custom antibody conjugation is available within the Immune Analysis Facility. Staining tissue with antibodies will be conducted within the facility or researcher’s lab.

Mass Cytometry Workflow
IMC Data Marker Visualization
IMC Data Multiplex
IsoPlex machine

The Isolight analyzer from Isoplexis is the newest addition to our facility.  This benchtop system performs automated highly mutliplexed live single cell secretome analysis, single cell highly multiplexed phosphoproteomic and metabolic analysis as well as bulk/population protein analysis all on one platform and in a single workflow. Fully analyzed data is received on the same day. IsoLight is ideal for comprehensive profiling of the secretory capabilities of different cell types and of cells at different developmental or treatment stages. Chips with different panels of analytes are available, for example:

  • Human Innate Immune IFN-γ, MIP-1α, TNF-α, TNF-β, GM-CSF, IL-8, IL-9, IL-15, IL-18, TGF-α, IL-5, CCL11, IP-10, MIP-1β, RANTES, BCA-1, IL-10, IL-13, IL-22, sCD40L, IL-1β, IL-6, IL-12-p40, IL-12, IL-17A, IL-17F, MCP-1, MCP-4, MIF, EGF, PDGF-BB, VEGF
  • Human Adaptive Immune Granzyme B, IFN-γ, MIP-1α, Perforin, TNF-α, TNF-β, GM-CSF, IL-2, IL-5, IL-7, IL-8, IL-9, IL-12, IL-15, IL-21*, CCL11, IP-10, MIP-1β, RANTES, IL-4, IL-10, IL-13, IL-22, TGFβ1, sCD137, sCD40L, IL-1β, IL-6, IL-17A, IL-17F, MCP-1, MCP-4
  • Human Inflammation GM-CSF, IFN-γ, IL-2, IL-12, TNF-α, TNF-β, IL-4, IL-5, IL-7, IL-9, IL-13, CCL11, IL-8, IP-10, MCP-1, MCP-4, MIP-1α, MIP-1β, RANTES, IL-10, IL-15, IL-22, TGF-β1, IL-1β, IL-6, IL-17A, IL-17F, IL-21*, Granzyme B, Perforin, sCD40L, sCD137
  • Human Natural Killer Granzyme B, IFN-γ, MIP-1α, Perforin, TNF-α, TNF-β, GM-CSF, IL-2, IL-5, IL-7, IL-8, IL-9, IL-12, IL-15, IL-21*, CCL11, IP-10, MIP-1β, RANTES, IL-4, IL-10, IL-13, IL-22, TGFβ1, sCD137, sCD40L, IL-1β, IL-6, IL-17A, IL-17F, MCP-1, MCP-4
  • Non-Human Primate Adaptive Immune TNF-α, MCP-1, IL-2, IL-4, MIP-1β, IL-6, IL-8, IL-1β, RANTES, IFN-g, IP-10, MIP-1α, MIF, GM-CSF
  • Mouse Innate Immune IFN-g, TNF-a, MIP-1a, IL-15, GM-CSF, IL-5, IL-10, IL-13, IL-6, IL-17A, MCP-1, IP-10, MIP-1b, EGF, PDGF-BB, MIF
  • Mouse Adaptive Immune Granzyme B, IFN-γ, MIP-1α, TNF-α, GM-CSF, IL-2, IL-5, IL-7, IL-12p70, IL-15, IL-21, sCD137, CCL11, CXCL1, CXCL13, IP-10, RANTES, Fas, IL-4, IL-10, IL-13, IL-27, TGFβ1, IL-6, IL-17A, MCP-1, IL-1β
Isolight Figure
Polyfunctional CTLs, i.e., CTLs capable of secreting more than one cytokine (e.g, granzyme-b, IFN-γ, MIP-1α, Perforin, TNFα and TNFβ) are often the best anti-tumor effector cells. IsoLight is ideal for identifying polyfunctional immune cells.

Equipment

Seahorse XFe96

Seahorse Xfe 96 Extracellular Flux Analyzer

CyTOF Helios mass cytometer

CyTOF Helios Mass Cytometer

Hyperion Imaging Mass Cytometer

Hyperion Imaging Mass Cytometer

CTL Core 6 ELISPOT plate reader

CTL-ImmunoSpot ELISPOT

Biotek Cytation 5

Bio-Tek Cytation 5

Synergy H1

BioTek Synergy H1 & HT Microplate Readers

ELLA, automated ELISA system

ELLA, Automated ELISA System

Luminex MAGPIX

Luminex MAGPIX System

Isolight machine

Isolight

Roswell Park Comprehensive Cancer Center

Location & hours

Roswell Park Comprehensive Cancer Center
Immune Analysis Facility
Cancer Cell Center, 4th Floor, Room 403 and 410
Elm and Carlton Streets
Buffalo, New York 14263

Monday – Friday, 8 a.m. – 4:30 p.m.

The Immune Analysis Facility is responsible for serial monitoring of immunologic functions in patients with cancer, patients treated with biologic therapies, and people participating in clinical trials or research protocols at Roswell Park or other outside institutions or hospitals. The facility makes available to its users a broad range of state-of-the-art immunologic assays, performed under a rigorous quality control program.

In addition, as advances in immunobiology occur and new assays are requested by the users, the IAF performs pre-clinical evaluations of the assays and, when they become reliable and standardized, adds them to the available assay list.

The IAF offers consultation regarding optimal immunologic assessment and assay development for innovative approaches to evaluate immune responses. We provide expert advice regarding the types of assays for immune monitoring and data interpretation tailored to fit the endpoints of each specific clinical trial.

The IAF maintains extensive quality control (QC) and quality assurance (QA) programs to ensure the validity of test results.

  • Choi JE, MacDonald CR, Gandhi N, Das G, Repasky EA, Mohammadpour H. Isolation of human and mouse myeloid-derived suppressor cells for metabolic analysis. STAR Protoc. 2022 JUN 17; 3(2):101389. DOI: 10.1016/j.xpro.2022.101389. 2022 May 17. PubMed PMID: 35600927; PMCID: PMC9120228.
  • Odunsi K, Qian F, Lugade AA, Yu H, Geller MA, Fling SP, Kaiser JC, Lacroix AM, D'Amico L, Ramchurren N, Morishima C, Disis ML, Dennis L, Danaher P, Warren S, Nguyen VA, Ravi S, Tsuji T, Rosario S, Zha W, Hutson A, Liu S, Lele S, Zsiros E, McGray AJR, Chiello J, Koya R, Chodon T, Morrison CD, Putluri V, Putluri N, Mager DE, Gunawan R, Cheever MA, Battaglia S, Matsuzaki J. Metabolic adaptation of ovarian tumors in patients treated with an IDO1 inhibitor constrains antitumor immune responses. Sci Transl Med. 2022 Mar 16;14(636):eabg8402. doi: 10.1126/scitranslmed.abg8402. Epub 2022 Mar 16. PMID: 35294258
  • Clough E, Inigo J, Chandra D, Chaves L, Reynolds JL, Aalinkeel R, Schwartz SA, Khmaladze A, Mahajan SD. Mitochondrial Dynamics in SARS-COV2 Spike Protein Treated Human Microglia: Implications for Neuro-COVID. J Neuroimmune Pharmacol. 2021 DEC 01; 16(4):770-784. DOI: 10.1007/s11481-021-10015-6. 2021 Oct 2. PubMed PMID: 34599743; PMCID: PMC8487226.
  • Mohammadpour H, MacDonald CR, McCarthy PL, Abrams SI, Repasky EA. β2-adrenergic receptor signaling regulates metabolic pathways critical to myeloid-derived suppressor cell function within the TME. Cell Rep. 2021 OCT 26; 37(4):109883. DOI: 10.1016/j.celrep.2021.109883. PubMed PMID: 34706232; PMCID: PMC8601406.
  • Thaker PH, Bradley WH, Leath CA, Gunderson Jackson C, Borys N, Anwer K, Musso L, Matsuzaki J, Bshara W, Odunsi K, Alvarez RD. GEN-1 in Combination with Neoadjuvant Chemotherapy for Patients with Advanced Epithelial Ovarian Cancer: A Phase I Dose-escalation Study. Clin. Cancer Res.. 2021 OCT 15; 27(20):5536-5545. DOI: 10.1158/1078-0432.CCR-21-0360. 2021 Jul 29. PubMed PMID: 34326131.
  • Chen M, Brackett CM, Burdelya LG, Punnanitinont A, Patnaik SK, Matsuzaki J, Odunsi AO, Gudkov AV, Singh AK, Repasky EA, Gurova KV. Stimulation of an anti-tumor immune response with "chromatin-damaging" therapy. Cancer Immunol. Immunother.. 2021 JUL 01; 70(7):2073-2086. DOI: 10.1007/s00262-020-02846-8. 2021 Jan 13. PubMed PMID: 33439292; PMCID: PMC8726059.
  • Emmons TR, Giridharan T, Singel KL, Khan ANH, Ricciuti J, Howard K, Silva-Del Toro SL, Debreceni IL, Aarts CEM, Brouwer MC, Suzuki S, Kuijpers TW, Jongerius I, Allen LH, Ferreira VP, Schubart A, Sellner H, Eder J, Holland SM, Ram S, Lederer JA, Eng KH, Moysich KB, Odunsi K, Yaffe MB, et al. Mechanisms Driving Neutrophil-Induced T-cell Immunoparalysis in Ovarian Cancer. Cancer Immunol Res. 2021 JUL 01; 9(7):790-810. DOI: 10.1158/2326-6066.CIR-20-0922. 2021 May 14. PubMed PMID: 33990375; PMCID: PMC8287091.
  • Gandhi S, Oshi M, Murthy V, Repasky EA, Takabe K. Enhanced Thermogenesis in Triple-Negative Breast Cancer Is Associated with Pro-Tumor Immune Microenvironment. Cancers (Basel). 2021 MAY 23; 13(11). DOI: 10.3390/cancers13112559. 2021 May 23. PubMed PMID: 34071012; PMCID: PMC8197168.
  • Chouliaras K, Tokumaru Y, Asaoka M, Oshi M, Attwood KM, Yoshida K, Ishikawa T, Takabe K. Prevalence and clinical relevance of tumor-associated tissue eosinophilia (TATE) in breast cancer. Surgery. 2021 MAY 01; 169(5):1234-1239. DOI: 10.1016/j.surg.2020.07.052. 2020 Sep 19. PubMed PMID: 32958266; PMCID: PMC7969474.
  • Ibrahim OM, Basse PH, Jiang W, Guru K, Chatta G, Kalinski P. NFκB-Activated COX2/PGE2/EP4 Axis Controls the Magnitude and Selectivity of BCG-Induced Inflammation in Human Bladder Cancer Tissues. Cancers (Basel). 2021 MAR 16; 13(6). DOI: 10.3390/cancers13061323. 2021 Mar 16. PubMed PMID: 33809455; PMCID: PMC7998891.
  • Yamauchi T, Hoki T, Oba T, Jain V, Chen H, Attwood K, Battaglia S, George S, Chatta G, Puzanov I, Morrison C, Odunsi K, Segal BH, Dy GK, Ernstoff MS, Ito F. T-cell CX3CR1 expression as a dynamic blood-based biomarker of response to immune checkpoint inhibitors. Nat Commun. 2021 MAR 03; 12(1):1402. DOI: 10.1038/s41467-021-21619-0. 2021 Mar 3. PubMed PMID: 33658501; PMCID: PMC7930182.
  • Want MY, Tsuji T, Singh PK, Thorne JL, Matsuzaki J, Karasik E, Gillard B, Cortes Gomez E, Koya RC, Lugade A, Odunsi K, Battaglia S. WHSC1/NSD2 regulates immune infiltration in prostate cancer. J Immunother Cancer. 2021 FEB 01; 9(2). DOI: 10.1136/jitc-2020-001374. PubMed PMID: 33589522; PMCID: PMC7887377.
  • Amobi-McCloud A, Muthuswamy R, Battaglia S, Yu H, Liu T, Wang J, Putluri V, Singh PK, Qian F, Huang RY, Putluri N, Tsuji T, Lugade AA, Liu S, Odunsi K. IDO1 Expression in Ovarian Cancer Induces PD-1 in T Cells via Aryl Hydrocarbon Receptor Activation. Front Immunol. 2021 JAN 01; 12:678999. DOI: 10.3389/fimmu.2021.678999. 2021 Apr 16. PubMed PMID: 34025677; PMCID: PMC8136272.