CD8+ cytotoxic T cells can be harvested from a patient with

Similar documents
NIH Public Access Author Manuscript Science. Author manuscript; available in PMC 2008 March 12.

Supporting Information

CONTRACTING ORGANIZATION: Johns Hopkins University School of Medicine Baltimore, MD 21205

Tumor responses (patients responding/ patients treated)

The Immune System. Innate. Adaptive. - skin, mucosal barriers - complement - neutrophils, NK cells, mast cells, basophils, eosinophils

DEVELOPMENT OF CELLULAR IMMUNOLOGY

CONTRACTING ORGANIZATION: Johns Hopkins University School of Medicine Baltimore, MD 21205

08/02/59. Tumor Immunotherapy. Development of Tumor Vaccines. Types of Tumor Vaccines. Immunotherapy w/ Cytokine Gene-Transfected Tumor Cells

Tumor Immunology. Wirsma Arif Harahap Surgical Oncology Consultant

COURSE: Medical Microbiology, PAMB 650/720 - Fall 2008 Lecture 16

Adoptive Cellular Therapy SITC Primer October 2012

Endogenous and Exogenous Vaccination in the Context of Immunologic Checkpoint Blockade

TITLE: Development of Antigen Presenting Cells for adoptive immunotherapy in prostate cancer

Madhav V. Dhodapkar, Joseph Krasovsky, Ralph M. Steinman, and Nina Bhardwaj

Chapter 7 Conclusions

Adoptive T Cell Therapy:

Third line of Defense

Determinants of Immunogenicity and Tolerance. Abul K. Abbas, MD Department of Pathology University of California San Francisco

Cancer Immunotherapy: Active Immunization Approaches

IMMUNOTHERAPY FOR CANCER A NEW HORIZON. Ekaterini Boleti MD, PhD, FRCP Consultant in Medical Oncology Royal Free London NHS Foundation Trust

Immunotherapy on the Horizon: Adoptive Cell Therapy

Exploring Immunotherapies: Beyond Checkpoint Inhibitors

TITLE: Development of Antigen Presenting Cells for adoptive immunotherapy in prostate cancer

Accepted Manuscript. Next Steps for Immune Checkpoints in Hepatocellular Carcinoma. Patricia M. Santos, Lisa H. Butterfield

Foundations in Microbiology

Cancer immunity and immunotherapy. General principles

Enhancing the Clinical Activity of HER2/neu Specific T Cells. William Gwin, MD Internal Medicine, Resident University of Washington

Kinetics of tumor-specific T-cell response development after active immunization in patients with HER-2/neu overexpressing cancers

Releasing the Brakes on Tumor Immunity: Immune Checkpoint Blockade Strategies

Interleukin-2 Single Agent and Combinations

TITLE: Development of Antigen Presenting Cells for adoptive immunotherapy in prostate cancer

Tumor Immunity and Immunotherapy. Andrew Lichtman M.D., Ph.D. Brigham and Women s Hospital Harvard Medical School

Synergistic combinations of targeted immunotherapy to combat cancer

Tumors arise from accumulated genetic mutations. Tumor Immunology (Cancer)

TITLE: MODULATION OF T CELL TOLERANCE IN A MURINE MODEL FOR IMMUNOTHERAPY OF PROSTATIC ADENOCARCINOMA

CHAPTER 18: Immune System

M.Sc. III Semester Biotechnology End Semester Examination, 2013 Model Answer LBTM: 302 Advanced Immunology

MHC class I MHC class II Structure of MHC antigens:

Cytotoxicity assays. Rory D. de Vries, PhD 1. Viroscience lab, Erasmus MC, Rotterdam, the Netherlands

ANTIBODY IMMUNITY TO PROSTATE CANCER ASSOCIATED ANTIGENS CAN BE DETECTED IN THE SERUM OF PATIENTS WITH PROSTATE CANCER

LAMPvax DNA Vaccines as Immunotherapy for Cancer - Three Case Studies

Principles of Adaptive Immunity

STATE OF THE ART 4: Combination Immune Therapy-Chemotherapy. Elizabeth M. Jaffee (JHU) James Yang (NCI) Jared Gollob (Duke) John Kirkwood (UPMI)

Chapter 22: The Lymphatic System and Immunity

Antigen Presentation and T Lymphocyte Activation. Abul K. Abbas UCSF. FOCiS

Tumor Antigens in the Age of Engineered T cell Therapies

Current Tumor Immunotherapy

Medical Virology Immunology. Dr. Sameer Naji, MB, BCh, PhD (UK) Head of Basic Medical Sciences Dept. Faculty of Medicine The Hashemite University

Micro 204. Cytotoxic T Lymphocytes (CTL) Lewis Lanier

DISCLOSURES. Roche/MSD-Merck/Celgene: Research Funding

LYMPHOCYTES & IMMUNOGLOBULINS. Dr Mere Kende, Lecturer SMHS

RAISON D ETRE OF THE IMMUNE SYSTEM:

The Major Histocompatibility Complex (MHC)

Immunology Lecture 4. Clinical Relevance of the Immune System

Third line of Defense. Topic 8 Specific Immunity (adaptive) (18) 3 rd Line = Prophylaxis via Immunization!

Priming the Immune System to Kill Cancer and Reverse Tolerance. Dr. Diwakar Davar Assistant Professor, Melanoma and Phase I Therapeutics

Tumor Immunology: A Primer

Effector mechanisms of cell-mediated immunity: Properties of effector, memory and regulatory T cells

ACTIVATION AND EFFECTOR FUNCTIONS OF CELL-MEDIATED IMMUNITY AND NK CELLS. Choompone Sakonwasun, MD (Hons), FRCPT

Immuno-Oncology Therapies and Precision Medicine: Personal Tumor-Specific Neoantigen Prediction by Machine Learning

1. Overview of Adaptive Immunity

Oncolytic Immunotherapy: A Local and Systemic Antitumor Approach

Therapeutic efficacy of MUC1- specific CTL and CD137 costimulation. mammary cancer model. Pinku Mukherjee & Sandra Gendler

IL-12 enhances the generation of tumour antigen-specific Th1 CD4 T cells during ex vivo expansion

Adverse effects of Immunotherapy. Asha Nayak M.D

Artificial Antigen Presenting Cells as a Standardized Platform for Tumor Infiltrating Lymphocyte (TIL) expansion

Cloning and expansion of antigen-specific T cells using ipsc technology: A novel strategy for cancer immunotherapy

Adaptive Immunity: Specific Defenses of the Host

Adoptive cell therapy using genetically modified antigen-presenting cells

Developing Novel Immunotherapeutic Cancer Treatments for Clinical Use

Adoptive Cell Therapy: Treating Cancer

New insights into CD8+ T cell function and regulation. Pam Ohashi Princess Margaret Cancer Centre

SUPPLEMENTARY INFORMATION

The NY-ESO-1 Ag is expressed by many tumors of different

Emerging Targets in Immunotherapy

Immuno-Oncology Therapies and Precision Medicine: Personal Tumor-Specific Neoantigen Prediction by Machine Learning

Immunotherapy for the Treatment of Cancer

IMMUNOTHERAPY IN THE TREATMENT OF CERVIX CANCER

Mucosal Immune System

General Overview of Immunology. Kimberly S. Schluns, Ph.D. Associate Professor Department of Immunology UT MD Anderson Cancer Center

Transplantation. Immunology Unit College of Medicine King Saud University

PBMC from each patient were suspended in AIM V medium (Invitrogen) with 5% human

The Good, the Bad and the Ugly: Clinical trials which assess vaccine characteristics. ISBT Meeting, San Francisco, CA November 4-8, 2004

Immune surveillance hypothesis (Macfarlane Burnet, 1950s)

Therapeutic Cancer Vaccines

T cell manipulation of the graft: Yes

Dr. Yi-chi M. Kong August 8, 2001 Benjamini. Ch. 19, Pgs Page 1 of 10 TRANSPLANTATION

chapter 17: specific/adaptable defenses of the host: the immune response

T-Cell Transfer Therapy Targeting Mutant KRAS in Cancer

T-cell activation T cells migrate to secondary lymphoid tissues where they interact with antigen, antigen-presenting cells, and other lymphocytes:

T-cell activation T cells migrate to secondary lymphoid tissues where they interact with antigen, antigen-presenting cells, and other lymphocytes:

Lecture outline. Immunological tolerance and immune regulation. Central and peripheral tolerance. Inhibitory receptors of T cells. Regulatory T cells

Immunological Tolerance

Cancer Immunotherapy. What is it? Immunotherapy Can Work! 4/15/09. Can the immune system be harnessed to fight cancer? T CD4 T CD28.

VISTA, a novel immune checkpoint protein ligand that suppresses anti-tumor tumor T cell responses. Li Wang. Dartmouth Medical School

Mon, Wed, Fri 11:00 AM-12:00 PM. Owen, Judy, Jenni Punt, and Sharon Stranford Kuby-Immunology, 7th. Edition. W.H. Freeman and Co., New York.

Endeavour College of Natural Health endeavour.edu.au

Melanocyte Destruction after Antigen-specific Immunotherapy of Melanoma: Direct Evidence of T Cell mediated Vitiligo

The Immune System. These are classified as the Innate and Adaptive Immune Responses. Innate Immunity

Immunologic and therapeutic evaluation of a synthetic peptide vaccine for the treatment of patients with metastatic melanoma

Transcription:

The new england journal of medicine brief report Treatment of Metastatic Melanoma with Autologous CD4+ T Cells against NY-ESO-1 Naomi N. Hunder, M.D., Herschel Wallen, M.D., Jianhong Cao, Ph.D., Deborah W. Hendricks, B.Sc., John Z. Reilly, B.Sc., Rebecca Rodmyre, B.Sc., Achim Jungbluth, M.D., Sacha Gnjatic, Ph.D., John A. Thompson, M.D., and Cassian Yee, M.D. Summary We developed an in vitro method for isolating and expanding autologous CD4+ T-cell clones with specificity for the melanoma-associated antigen NY-ESO-1. We infused these cells into a patient with refractory metastatic melanoma who had not undergone any previous conditioning or cytokine treatment. We show that the transferred CD4+ T cells mediated a durable clinical remission and led to endogenous responses against melanoma antigens other than NY-ESO-1. From the Fred Hutchinson Cancer Research Center (N.N.H., H.W., J.C., D.W.H., J.Z.R., R.R., C.Y.) and the University of Washington (J.A.T., C.Y.) both in Seattle; and the Ludwig Institute for Cancer Research (New York Branch), Memorial Sloan-Kettering Cancer Center, New York (A.J., S.G.). Address reprint requests to Dr. Yee at the Fred Hutchinson Cancer Research Center, Room D3-1, 11 Fairview Ave. N., Seattle, WA 9819, or at cyee@fhcrc.org. N Engl J Med 28;358:2698-73. Copyright 28 Massachusetts Medical Society. CD8+ cytotoxic T cells can be harvested from a patient with cancer, expanded in vitro, selected for specificity against a tumor-associated antigen, and infused back into the patient. Such autologous T cells have been shown in clinical trials to have a beneficial effect in some patients with cancer. 1-5 The cytotoxic antitumor effect of CD8+ T cells depends on CD4+ T cells, which provide CD8+ T cells with growth factors such as interleukin-2 and can mediate the destruction of tumor cells either directly or indirectly. 5-8 Growth factors such as interleukin-2 can act in an autocrine manner, which in principle would allow an infusion of CD4+ T cells to proliferate in the patient and stimulate endogenous antitumor CD8+ T cells. Until recently, however, a means of isolating and expanding antitumor CD4+ T cells in numbers sufficient for cellular therapy has not been feasible. The identification of HLA class II restricted epitopes in tumorassociated antigens such as NY-ESO-1 and tyrosinase and the development of methods for the isolation and expansion of CD4+ T cells in vitro gave us the opportunity to implement a clinical trial to evaluate the safety, in vivo persistence, and antitumor efficacy of autologous CD4+ T-cell clones for the treatment of metastatic melanoma. We describe a patient with refractory metastatic melanoma who had a long-term complete remission after receiving autologous NY-ESO-1 specific CD4+ T-cell clones. Case Report The patient was 52 years old when he presented with recurrent melanoma and pulmonary and left iliac and inguinal metastases. His tumor had not responded to high-dose interferon alfa, four cycles of high-dose interleukin-2, and local excision. Baseline staging with magnetic resonance imaging of the brain and computed tomography (CT) of the chest, abdomen, and pelvis showed metastatic disease in the posterior right pleura, right hilum, and left iliac region. There were no central ner- 2698 n engl j med 358;25 www.nejm.org june 19, 28

Brief Report vous system metastases. Positron-emission tomography (PET) of the entire body revealed no other sites of uptake. Immunohistochemical analysis that was performed on a biopsy specimen of the inguinal tumor revealed uniform expression of melanoma antigen recognized by T cells (MART-1, also called melanoma tumor antigen [MelanA]), 3+ staining of melanoma antigen 3 (MAGE-3), 3+ staining of NY-ESO-1, and no detectable glycoprotein 1. The patient was enrolled in a clinical trial of autologous T-cell therapy for melanoma at the Fred Hutchinson Cancer Research Center in Seattle (protocol 1585). The study was approved by the institutional review board, and the patient provided written informed consent. Peripheral-blood mononuclear cells were collected by leukopheresis, and T cells in the harvest were cultured. Since the patient s HLA genotype included the HLA-DOB1*41 allele, CD4+ T-cell clones targeting the DPB1*41-restricted epitope of a peptide derived from NY-ESO-1 were isolated from the culture and expanded for cellular therapy. In vitro, these clones responded to the NY-ESO-1 peptide by producing interleukin-2 and interferon-γ. During a 2-hour period, the patient received a single infusion of 3.3 1 9 clonal CD4+ T cells per square meter of body-surface area, totaling 5 billion T cells. No preinfusion conditioning or postinfusion interleukin-2 was administered. Transient lymphopenia, low-grade fever (maximum temperature, <38.5 C), and myalgia, all consistent with cytokine release, 3 developed and resolved within 3 days after the infusion. There were no serious adverse events, according to the Common Toxicity Criteria of the National Cancer Institute. Two months after the infusion, restaging PET and CT scans revealed complete resolution of pulmonary and nodal disease; there was no evidence of abnormal fluorodeoxyglucose uptake and no other radiographic or clinical evidence of disease (Fig. 1). The patient remained disease-free 2 years later. Methods Before Infusion After Infusion Pulmonary Nodule Generation of Antigen-Specific CD4+ T-Cell Clones Antigen-specific CD4+ T-cell clones were generated in a manner similar to that for CD8+ T cells described previously. 3,9 Brief ly, autologous monocyte-derived dendritic cells were pulsed with 4 μg of the NY-ESO-1 peptide SLLMWITQCFLPVF 1 per milliliter and cocultured with the patient s T cells at a ratio of 1:1 (T cells:dendritic cells). Interleukin-2 (12.5 IU per milliliter) and interleukin-7 (1 ng per milliliter) were added every 3 days to maintain the growth of activated antigenspecific T cells. One week later, the T cells were restimulated with peptide-pulsed autologous mononuclear cells. One week after the second stimulation, T cells with specific reactivity against the NY-ESO-1 peptide were harvested and cloned at limiting dilution in the presence of irradiated feeder cells (mononuclear cells plus Epstein Barr virus transformed B cells), a monoclonal anti-cd3 antibody, and interleukin-2. Clones that showed a specific proliferative response to the NY-ESO-1 peptide in a thymidine incorporation assay were expanded in vitro. A total of four NY-ESO-1 specific CD4+ T-cell clones were generated from the patient. The clone that was selected for infusion could be expanded by a factor of 3 to 5 during a period of 18 days with the use of established methods. 11 Reactivity against NY-ESO-1 was confirmed after expansion by performing assays to detect antigen-specific proliferation and the re- Inguinal Nodule Figure 1. PET Scans Obtained before T-Cell Infusion and 2 Months after Infusion. Preinfusion positron-emission tomography (PET) reveals hypermetabolic lesions in the right lung and the left inguinal iliac node that are consistent with metastases (arrowheads); the lesions colocalized to tumor nodules on computed tomography (CT). After infusion with NY-ESO-1 specific CD4+ T cells, PET scans show that the tumor nodules have regressed, and no evidence of disease can be detected. Uptake in liver, spleen, and bladder represents normal background signal and did not colocalize to any lesions on CT scans. n engl j med 358;25 www.nejm.org june 19, 28 2699

The new england journal of medicine lease of interferon-γ (for details, see the Supplementary Appendix, available with the full text of this article at www.nejm.org). Detection of Antibodies against Tumor-Associated Antigens An enzyme-linked immunosorbent assay was used to detect IgG antibodies against recombinant fulllength NY-ESO-1, MAGE-3, and MART-1 proteins (1 μg per milliliter), as described previously. 12 Reciprocal titers were extrapolated from serial dilutions, and titers of more than 1 were considered to be reactive. Specificity was determined by comparing reactivity with that of control antigens. Assay for Antigen-Specific Responses To evaluate the postinfusion T-cell responses to a panel of melanoma antigens, RNA-transfected autologous dendritic cells were used as stimulator cells in enzyme-linked immunospot (ELISPOT) assays. Melanoma antigens MART-1, glycoprotein 1, NY-ESO-1, and MAGE-3 were subcloned into the pgem4z vector, flanked by the signal sequence from lysosomal-associated membrane protein 1 (LAMP-1) to facilitate antigen sorting to both class I and class II antigen-presentation pathways (for details, see the Supplementary Appendix). 13 For the inteferon-γ ELISPOT assay, the patient s mononuclear cells were cocultivated with RNAtransfected dendritic cells at a ratio of 2:1 in triplicate filter wells. After overnight incubation, the plates were treated with a biotinylated anti interferon-γ-detection antibody, clone 7B61 (Mabtech). Streptavidin HRP (Pharmingen) was added and developed to produce a color reaction with the use of the Vectastain AEC substrate kit (Vector Laboratories). Each spot on the filter well represented a single antigen-stimulated T cell. Spots were enumerated and the antigen-specific T-cell frequency was calculated as a fraction of the input number of mononuclear cells (see the Supplementary Appendix for details). T-Cell Tracking by Quantitative PCR We detected cells of the NY-ESO-1 specific CD4+ T-cell clone in the patient s blood using primers flanking the CDR3 region of the T-cell receptor of the clone. Genomic DNA from mononuclear cells was harvested and used as a template for a quantitative real-time polymerase-chain-reaction (PCR) assay. The frequency of the cells was determined by reconciling the melting temperature of the sample s reaction with that obtained from standards titrated with known frequencies of the CD4+ T-cell clone. This assay can detect a T-cell clone in peripheral blood at a sensitivity of 1:1, cells. Results Persistence of the CD4+ T-Cell Clone The persistence of the infused NY-ESO-1 specific CD4+ T-cell clone in vivo was evaluated with the use of a quantitative PCR assay that can detect the clone-specific CDR3 region in samples of mononuclear cells. The T-cell clone was undetectable in preinfusion samples from the patient. After infusion, however, there was a rapid rise in the frequency of cells of the clone: they constituted almost 2.% of all peripheral-blood mononuclear cells on day 3 and remained detectable in the patient s blood for more than 8 days (Fig. 2). During this time, the frequency of cells of the clone fluctuated between.7 and 3.% of the total number of mononuclear cells. Clinical Evaluation CT and PET scans obtained 2 months after the T-cell infusion revealed no evidence of disease, and follow-up after 22 months showed no evidence of recurrence (Fig. 1). At the time of the last contact with the patient, 26 months after the T-cell infusion, he had received no other treatment and had normal function, with a Karnofsky performance status of 9 to 1%. There were no detectable acute or long-term autoimmune-related toxic effects (e.g., dermatitis, vitiligo, uveitis, and orchitis). T-Cell Responses to Unrelated Antigens Even though only 5 to 75% of the tumor cells expressed NY-ESO-1, the entire tumor regressed after infusion of the NY-ESO-1 specific CD4+ T-cell clone. This inconsistency led us to postulate that the infused clone induced an immune response that was broader than expected. To test this hypothesis, we evaluated T-cell responses to two other melanoma-associated antigens that were expressed by the patient s tumor, MART-1 and MAGE-3. Using autologous dendritic cells that were engineered by RNA transfection to express these antigens, we could detect T cells in the patient s blood that responded in vitro to MART-1 or MAGE-3. These T cells were undetectable before the infusion of NY-ESO-1 specific CD4+ T cells 27 n engl j med 358;25 www.nejm.org june 19, 28

Brief Report 35, No. of CD4+ T-Cell Clones per 1 6 3, 25, 2, 15, 1, 5, 1.9%.7% 3.% 1.% 1 2 3 4 5 6 7 8 9 1 Figure 2. Persistence of Transferred NY-ESO-1 Specific CD4+ T-Cell Clones In Vivo. Quantitative polymerase-chain-reaction (PCR) assay with the use of clone-specific primers flanking the TCR CDR3 region was used to determine the frequency of infused CD4+ T-cell clones among peripheral-blood mononuclear cells () in samples collected before infusion and at intervals after infusion until day 9. Results are represented as the number of T-cell clones per 1 million and as a percentage of the total number of at peak points and low points. As expected, the NY-ESO-1 specific CD4+ T-cell clone is undetectable in preinfusion (sensitivity of detection, 1:1, cells). After infusion, the frequency of the infused CD4+ T-cell clone in vivo increased to almost 2% of all and fluctuated between.7 and 3.% during the next 12 weeks. The I bars denote standard errors. but became detectable at a time that coincided with regression of the tumor and persisted for at least 3 months. We found no evidence of T cells that could respond to glycoprotein 1, which was poorly expressed by the tumor (Fig. 3). Discussion We describe an in vitro method for the development of CD4+ T-cell clones with specificity for a tumor antigen and the use of one such clone for the treatment of a patient with metastatic melanoma. Development of the method has been difficult, in part because of the paucity of well-defined immunogenic HLA class II restricted epitopes of tumor antigens and also because of the difficulty in isolating and expanding CD4+ T-cell clones to obtain numbers sufficient for clinical use. The HLA-DP4 restricted peptide epitope of the melanoma-associated cancer testis antigen NY-ESO-1 is one of the most immunogenic tumor antigens 1,14 and can elicit responses by CD4+ T cells from patients with cancer who have been immunized with NY-ESO-1. 15,16 Using the NY-ESO-1 peptide, we succeeded in generating NY-ESO-1 specific CD4+ T cells from the blood of an unimmunized patient with metastatic melanoma. We were able to expand these cells to several billion and use them to treat the patient. No exogenous cytokines were administered because these CD4+ T cells, when stimulated in vitro by the NY-ESO-1 antigen, produce autocrine-active interleukin-2. After infusion of the cells, influenza-like symptoms and lymphopenia, signs of a cytokine release syndrome, developed. In contrast to transferred CD8+ T cells, which survive only briefly (<2 days) in vivo in the absence of exogenous cytokine, 1,3 the CD4+ T-cell clones that we administered without any cytokine treatment were detectable in the patient s blood for at least 3 months. Therefore, the possibility that in vitro derived and clonally expanded T cells were functionally exhausted 17 was not substantiated by our results. Our studies and those performed by others have shown that immunotherapy with antigenspecific T cells can eliminate tumor cells that express the corresponding antigen, but it can also allow the outgrowth of antigen-loss tumor variants. 1,3 The target antigen in our case, NY-ESO-1, was not uniformly expressed by the cells of the patient s melanoma, yet the tumor regressed com- n engl j med 358;25 www.nejm.org june 19, 28 271

The new england journal of medicine A MAGE-3 No. of Spots per 1, C NY-ESO-1 No. of Spots per 1, 14 12 DCs with MAGE-3 RNA 1 8 6 4 2 DCs not transfected with RNA 1 2 3 4 5 6 7 8 9 25 2 15 1 5 DCs not transfected with RNA 1 2 3 4 5 6 7 8 9 DCs with NY-ESO-1 RNA B MART-1 No. of Spots per 1, D GP1 No. of Spots per 1, 18 16 14 12 1 8 DCs with MART-1 RNA 6 4 DCs not transfected with RNA 2 1 2 3 4 5 6 7 8 9 1 8 6 4 2 DCs not transfected with RNA DCs with GP1 RNA 1 2 3 4 5 6 7 8 9 Figure 3. Augmentation of T-Cell Responses to Melanoma-Associated Tumor Antigens. Antigen-specific T-cell responses to melanoma antigen recognized by T cells (MART-1), melanoma antigen 3 (MAGE-3), NY-ESO-1, and glycoprotein 1 (GP1) were evaluated in samples of peripheral-blood mononuclear cells () collected before infusion and at time points up to 12 weeks after infusion. Autologous dendritic cells (DCs) that were transfected with RNA-encoding tumor antigen were used to stimulate in enzyme-linked immunospot assays. The frequency of the antigen-specific response is presented as the number of reactive spots per 1, (squares). Parallel wells with DCs transfected with control RNA were used at each time point (diamonds). Each data point is an average of the values from triplicate wells. After infusion, marked elevations in antigen-specific T-cell responses were observed for MAGE-3 (Panel A), MART-1 (Panel B), and NY-ESO-1 (Panel C) but not for GP1 (Panel D). The I bars denote standard errors. pletely. We postulate that the complete regression of the tumor can be attributed in part to responses by the patient s immune system to other antigens displayed by the tumor. This additional response could have come about through antigens in apoptotic bodies that were released from killed tumor cells. When collected and processed by antigen-presenting cells, these antigens probably activated a variety of T cells, thereby broadening the immune response, a process referred to as antigen spreading. 18,19 The delay of days to weeks in the development of these additional responses is consistent with antigen spreading (Fig. 3). The fact that T cells against glycoprotein 1, an antigen that was not expressed by the melanoma, were undetectable indicates the selectivity of the antitumor immune responses. It is also possible that the reduction in the tumor burden relieved immunosuppression in the tumor microenvironment caused by inhibitory cytokines and regulatory T cells, thereby contributing to the broadened T-cell response. IgG antibodies against NY-ESO-1 were detectable in plasma samples from the patient before infusion of the T-cell clone and remained unchanged in titer after the infusion. The presence of antibodies against NY-ESO-1 is associated with an increased likelihood that NY-ESO-1 specific CD4+ and CD8+ T-cell responses will be generated ex vivo. 14,2 Antibodies against MAGE-3 and MART-1 were undetectable before the T-cell infusion and afterward for up to 87 days (Fig. 1 in the Supplementary Appendix). The lack of an increase in the titers of antibodies against melanoma antigens despite the appearance of CD4+ T cells against these antigens suggests that the production of such antibodies is independent of T cells. In summary, we showed that the infusion of a clonal population of CD4+ T cells with specificity for a single tumor-associated antigen caused complete regression of a tumor. During regression of the tumor, this clone appears to have induced 272 n engl j med 358;25 www.nejm.org june 19, 28

Brief Report the patient s own T cells to respond to other antigens of his tumor. These findings support further clinical studies of antigen-specific CD4+ T cells in the treatment of malignant disease. No potential conflict of interest relevant to this article was reported. Supported in part by grants from the National Institutes of Health (CA12823 and CA147113) and the General Clinical Research Center (M1-RR-37) and by the Edson Foundation and the Damon Runyon Cancer Research Foundation. Dr. Yee is a recipient of the Clinical Scientist Award in Translational Research from the Burroughs Wellcome Fund. We thank Yong Qing Li and Kaye Dowdy for their technical assistance, Lisa Schirmer for assistance in clinical research support, and Sandra Gan for logistical support. References 1. Mackensen A, Meidenbauer N, Vogl S, Laumer M, Berger J, Andreesen R. Phase I study of adoptive T-cell therapy using antigen-specific CD8+ T cells for the treatment of patients with metastatic melanoma. J Clin Oncol 26;24:56-9. 2. Mitchell MS, Darrah D, Yeung D, et al. Phase I trial of adoptive immunotherapy with cytolytic T lymphocytes immunized against a tyrosinase epitope. J Clin Oncol 22;2:175-86. 3. Yee C, Thompson JA, Byrd D, et al. Adoptive T cell therapy using antigen-specific CD8+ T cell clones for the treatment of patients with metastatic melanoma: in vivo persistence, migration, and antitumor effect of transferred T cells. Proc Natl Acad Sci U S A 22;99:16168-73. 4. Dudley ME, Wunderlich JR, Robbins PF, et al. Cancer regression and autoimmunity in patients after clonal repopulation with antitumor lymphocytes. Science 22;298:85-4. 5. Greenberg PD. Adoptive T cell therapy of tumors: mechanisms operative in the recognition and elimination of tumor cells. Adv Immunol 1991;49:281-355. 6. Romerdahl CA, Kripke ML. Role of helper T-lymphocytes in rejection of UVinduced murine skin cancers. Cancer Res 1988;48:2325-8. 7. Hung K, Hayashi R, Lafond-Walker A, Lowenstein C, Pardoll D, Levitsky H. The central role of CD4(+) T cells in the antitumor immune response. J Exp Med 1998; 188:2357-68. 8. Greenberg PD, Kern DE, Cheever MA. Therapy of disseminated murine leukemia with cyclophosphamide and immune Lyt-1+,2- T cells: tumor eradication does not require participation of cytotoxic T cells. J Exp Med 1985;161:1122-34. 9. Li Y, Bleakley M, Yee C. IL-21 influences the frequency, phenotype, and affinity of the CD8 T cell response. J Immunol 25;175:2261-9. 1. Zeng G, Li Y, El-Gamil M, et al. Generation of NY-ESO-1-specific CD4+ and CD8+ T cells by a single peptide with dual MHC class I and class II specificities: a new strategy for vaccine design. Cancer Res 22;62:363-5. 11. Riddell SR, Greenberg PD. The use of anti-cd3 and anti-cd28 monoclonal antibodies to clone and expand human antigen-specific T cells. J Immunol Methods 199;128:189-21. 12. Stockert E, Jäger E, Chen YT, et al. A survey of the humoral immune response of cancer patients to a panel of human tumor antigens. J Exp Med 1998;187:1349-54. 13. Liao X, Li Y, Bonini C, et al. Transfection of RNA encoding tumor antigens following maturation of dendritic cells leads to prolonged presentation of antigen and the generation of high-affinity tumor-reactive cytotoxic T lymphocytes. Mol Ther 24;9:757-64. 14. Gnjatic S, Atanackovic D, Jäger E, et al. Survey of naturally occurring CD4+ T cell responses against NY-ESO-1 in cancer patients: correlation with antibody responses. Proc Natl Acad Sci U S A 23;1: 8862-7. 15. Odunsi K, Qian F, Matsuzaki J, et al. Vaccination with an NY-ESO-1 peptide of HLA class I/II specificities induces integrated humoral and T cell responses in ovarian cancer. Proc Natl Acad Sci U S A 27;14:12837-42. 16. Khong HT, Yang JC, Topalian SL, et al. Immunization of HLA-A*21 and/or HLA-DPbeta1*4 patients with metastatic melanoma using epitopes from the NY- ESO-1 antigen. J Immunother 24;27: 472-7. 17. Gattinoni L, Powell DJ Jr, Rosenberg SA, Restifo NP. Adoptive immunotherapy for cancer: building on success. Nat Rev Immunol 26;6:383-93. 18. Nowak AK, Lake RA, Marzo AL, et al. Induction of tumor cell apoptosis in vivo increases tumor antigen cross-presentation, cross-priming rather than cross-tolerizing host tumor-specific CD8 T cells. J Immunol 23;17:495-13. 19. Bevan MJ. Cross-priming for a secondary cytotoxic response to minor H antigens with H-2 congenic cells which do not cross-react in the cytotoxic assay. J Exp Med 1976;143:1283-8. 2. Jäger E, Chen YT, Drijfhout JW, et al. Simultaneous humoral and cellular immune response against cancer-testis antigen NY-ESO-1: definition of human histocompatibility leukocyte antigen (HLA)-A2-binding peptide epitopes. J Exp Med 1998;187: 265-7. Copyright 28 Massachusetts Medical Society. posting presentations at medical meetings on the internet Posting an audio recording of an oral presentation at a medical meeting on the Internet, with selected slides from the presentation, will not be considered prior publication. This will allow students and physicians who are unable to attend the meeting to hear the presentation and view the slides. If there are any questions about this policy, authors should feel free to call the Journal s Editorial Offices. n engl j med 358;25 www.nejm.org june 19, 28 273