Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Cancer Discov ; 8(8): 935-943, 2018 08.
Article in English | MEDLINE | ID: mdl-29899062

ABSTRACT

A promising arsenal of targeted and immunotherapy treatments for metastatic melanoma has emerged over the last decade. With these therapies, we now face new mechanisms of tumor-acquired resistance. We report here a patient whose metastatic melanoma underwent dedifferentiation as a resistance mechanism to adoptive T-cell transfer therapy (ACT) to the MART1 antigen, a phenomenon that had been observed only in mouse studies to date. After an initial period of tumor regression, the patient presented in relapse with tumors lacking melanocytic antigens (MART1, gp100) and expressing an inflammation-induced neural crest marker (NGFR). We demonstrate using human melanoma cell lines that this resistance phenotype can be induced in vitro by treatment with MART1 T cell receptor-expressing T cells or with TNFα, and that the phenotype is reversible with withdrawal of inflammatory stimuli. This supports the hypothesis that acquired resistance to cancer immunotherapy can be mediated by inflammation-induced cancer dedifferentiation.Significance: We report a patient whose metastatic melanoma underwent inflammation-induced dedifferentiation as a resistance mechanism to ACT to the MART1 antigen. Our results suggest that future melanoma ACT protocols may benefit from the simultaneous targeting of multiple tumor antigens, modulating the inflammatory response, and inhibition of inflammatory dedifferentiation-inducing signals. Cancer Discov; 8(8); 935-43. ©2018 AACR.This article is highlighted in the In This Issue feature, p. 899.


Subject(s)
Drug Resistance, Neoplasm , MART-1 Antigen/immunology , Melanoma/therapy , Nerve Tissue Proteins/metabolism , Nevus, Pigmented/therapy , Receptors, Nerve Growth Factor/metabolism , Cell Dedifferentiation , Cell Line, Tumor , Coculture Techniques , Humans , Immunotherapy, Adoptive , Male , Melanoma/immunology , Middle Aged , Neoplasm Metastasis , Nevus, Pigmented/immunology , Receptors, Chimeric Antigen/metabolism , Recurrence
2.
Cancer Res ; 77(17): 4697-4709, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28652246

ABSTRACT

Evolution of tumor cell phenotypes promotes heterogeneity and therapy resistance. Here we found that induction of CD73, the enzyme that generates immunosuppressive adenosine, is linked to melanoma phenotype switching. Activating MAPK mutations and growth factors drove CD73 expression, which marked both nascent and full activation of a mesenchymal-like melanoma cell state program. Proinflammatory cytokines like TNFα cooperated with MAPK signaling through the c-Jun/AP-1 transcription factor complex to activate CD73 transcription by binding to an intronic enhancer. In a mouse model of T-cell immunotherapy, CD73 was induced in relapse melanomas, which acquired a mesenchymal-like phenotype. We also detected CD73 upregulation in melanoma patients progressing under adoptive T-cell transfer or immune checkpoint blockade, arguing for an adaptive resistance mechanism. Our work substantiates CD73 as a target to combine with current immunotherapies, but its dynamic regulation suggests limited value of CD73 pretreatment expression as a biomarker to stratify melanoma patients. Cancer Res; 77(17); 4697-709. ©2017 AACR.


Subject(s)
5'-Nucleotidase/metabolism , Gene Expression Regulation, Neoplastic , Immunotherapy , Inflammation/complications , Melanoma/pathology , Mitogen-Activated Protein Kinase 1/metabolism , T-Lymphocytes/transplantation , Adenosine/metabolism , Adoptive Transfer , Animals , GPI-Linked Proteins/metabolism , Humans , Inflammation/pathology , Melanoma/immunology , Melanoma/metabolism , Melanoma/therapy , Mice , Mice, Inbred C57BL , Neoplasm Invasiveness , Prognosis , Retrospective Studies , Transcription Factor AP-1/metabolism , Tumor Cells, Cultured
3.
Clin Cancer Res ; 23(16): 4556-4568, 2017 Aug 15.
Article in English | MEDLINE | ID: mdl-28468947

ABSTRACT

Purpose: A phase I study was conducted to determine safety, clinical efficacy, and antitumor immune responses in patients with advanced non-small cell lung carcinoma (NSCLC) following intratumoral administration of autologous dendritic cells (DC) transduced with an adenoviral (Ad) vector expressing the CCL21 gene (Ad-CCL21-DC). We evaluated safety and tumor antigen-specific immune responses following in situ vaccination (ClinicalTrials.gov: NCT01574222).Experimental Design: Sixteen stage IIIB/IV NSCLC subjects received two vaccinations (1 × 106, 5 × 106, 1 × 107, or 3 × 107 DCs/injection) by CT- or bronchoscopic-guided intratumoral injections (days 0 and 7). Immune responses were assessed by tumor antigen-specific peripheral blood lymphocyte induction of IFNγ in ELISPOT assays. Tumor biopsies were evaluated for CD8+ T cells by IHC and for PD-L1 expression by IHC and real-time PCR (RT-PCR).Results: Twenty-five percent (4/16) of patients had stable disease at day 56. Median survival was 3.9 months. ELISPOT assays revealed 6 of 16 patients had systemic responses against tumor-associated antigens (TAA). Tumor CD8+ T-cell infiltration was induced in 54% of subjects (7/13; 3.4-fold average increase in the number of CD8+ T cells per mm2). Patients with increased CD8+ T cells following vaccination showed significantly increased PD-L1 mRNA expression.Conclusions: Intratumoral vaccination with Ad-CCL21-DC resulted in (i) induction of systemic tumor antigen-specific immune responses; (ii) enhanced tumor CD8+ T-cell infiltration; and (iii) increased tumor PD-L1 expression. Future studies will evaluate the role of combination therapies with PD-1/PD-L1 checkpoint inhibition combined with DC-CCL21 in situ vaccination. Clin Cancer Res; 23(16); 4556-68. ©2017 AACR.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Carcinoma, Non-Small-Cell Lung/therapy , Chemokine CCL21/immunology , Dendritic Cells/immunology , Immunotherapy, Adoptive/methods , Lung Neoplasms/therapy , Adult , Aged , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , B7-H1 Antigen/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/immunology , Chemokine CCL21/genetics , Cohort Studies , Dendritic Cells/metabolism , Dendritic Cells/transplantation , Dyspnea/etiology , Female , Humans , Immunotherapy, Adoptive/adverse effects , Injections, Intralesional , Interferon-gamma/immunology , Interferon-gamma/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Male , Middle Aged , Muscle Weakness/etiology , Pain/etiology
4.
Cancer Immunol Res ; 5(5): 417-424, 2017 05.
Article in English | MEDLINE | ID: mdl-28411193

ABSTRACT

We explored the association between liver metastases, tumor CD8+ T-cell count, and response in patients with melanoma or lung cancer treated with the anti-PD-1 antibody, pembrolizumab. The melanoma discovery cohort was drawn from the phase I Keynote 001 trial, whereas the melanoma validation cohort was drawn from Keynote 002, 006, and EAP trials and the non-small cell lung cancer (NSCLC) cohort from Keynote 001. Liver metastasis was associated with reduced response and shortened progression-free survival [PFS; objective response rate (ORR), 30.6%; median PFS, 5.1 months] compared with patients without liver metastasis (ORR, 56.3%; median PFS, 20.1 months) P ≤ 0.0001, and confirmed in the validation cohort (P = 0.0006). The presence of liver metastasis significantly increased the likelihood of progression (OR, 1.852; P < 0.0001). In a subset of biopsied patients (n = 62), liver metastasis was associated with reduced CD8+ T-cell density at the invasive tumor margin (liver metastasis+ group, n = 547 ± 164.8; liver metastasis- group, n = 1,441 ± 250.7; P < 0.016). A reduced response rate and shortened PFS was also observed in NSCLC patients with liver metastasis [median PFS, 1.8 months; 95% confidence interval (CI), 1.4-2.0], compared with those without liver metastasis (n = 119, median PFS, 4.0 months; 95% CI, 2.1-5.1), P = 0.0094. Thus, liver metastatic patients with melanoma or NSCLC that had been treated with pembrolizumab were associated with reduced responses and PFS, and liver metastases were associated with reduced marginal CD8+ T-cell infiltration, providing a potential mechanism for this outcome. Cancer Immunol Res; 5(5); 417-24. ©2017 AACR.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Liver Neoplasms/drug therapy , Lung Neoplasms/drug therapy , Melanoma/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Adult , Aged , Aged, 80 and over , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/pathology , Disease-Free Survival , Female , Humans , Kaplan-Meier Estimate , Liver Neoplasms/immunology , Liver Neoplasms/secondary , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lymphocytes, Tumor-Infiltrating/immunology , Male , Melanoma/immunology , Melanoma/pathology , Middle Aged , Treatment Outcome , Young Adult
5.
Cancer Immunol Res ; 5(2): 118-126, 2017 02.
Article in English | MEDLINE | ID: mdl-28039162

ABSTRACT

Tumors expressing programmed death ligand 1 (PD-L1) interact with the corresponding negative-signal generating immune receptor on the surface of CD8 T cells, PD-1, thereby suppressing antitumor activity. Therapeutics blocking this interaction have shown promise in various cancers by restoring functional antitumor T-cell activity. We explored the degree of PD-L1, PD-1, and CD8 expression in a retrospective analysis of 29 clinical synovial sarcoma samples. Quantitative immunohistochemistry and multiplex immunofluorescence were used to determine relative quantification of CD8+ and PD-1+ T cells and PD-L1 expression within the intratumor area and the interface between the tumor and the surrounding nontumor tissue (i.e., invasive margin), and colocalization of these factors, respectively. PD-L1, PD-1, and CD8 cell densities in the tumor-invasive margins were significantly higher in the metastatic tumors than the primary tumors (P < 0.01), and PD-L1, PD-1, and CD8 cell densities were all significantly positively correlated with one other (P < 0.0001). PD-1 cell density in the tumor-invasive margin was significantly associated with worse progression-free survival. Multiplex immunofluorescence demonstrated coexpression of PD-1 and CD8 on lymphocytes within the invasive margin, as well as relative proximity between PD-1+ CD8 cells and PD-L1+ tumor cells. Our results provide a preclinical rationale for screening of patients with synovial sarcoma for the colocalization of CD8, PD-1, and PD-L1, which may be a marker for response to PD-1 blockade therapy. Cancer Immunol Res; 5(2); 118-26. ©2016 AACR.


Subject(s)
B7-H1 Antigen/genetics , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Programmed Cell Death 1 Receptor/genetics , Sarcoma, Synovial/genetics , Sarcoma, Synovial/immunology , B7-H1 Antigen/metabolism , Gene Expression , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Neoplasm Metastasis , Neoplasm Staging , Programmed Cell Death 1 Receptor/metabolism , Retrospective Studies , Sarcoma, Synovial/diagnosis , Sarcoma, Synovial/mortality
6.
BJR Case Rep ; 3(1): 20160002, 2017.
Article in English | MEDLINE | ID: mdl-30363303

ABSTRACT

Ipilimumab is a monoclonal cytotoxic T-lymphocyte-associated protein 4 antibody that has demonstrated improved survival in cutaneous melanoma. Little is known about the clinical impact of combining anti-cytotoxic T-lymphocyteassociated protein 4 therapy with radiation. Here we report a case of severe cutaneous desquamation in a 70-year-old female with vaginal/vulvar melanoma receiving concurrent ipilimumab and radiation therapy. The toxicity was successfully treated with oral/topical steroids and a break from treatment. This case underscores the importance of future research on optimal strategies for combining radiation with novel anti-tumour agents.

7.
Cancer Discov ; 7(2): 188-201, 2017 02.
Article in English | MEDLINE | ID: mdl-27903500

ABSTRACT

Loss-of-function mutations in JAK1/2 can lead to acquired resistance to anti-programmed death protein 1 (PD-1) therapy. We reasoned that they may also be involved in primary resistance to anti-PD-1 therapy. JAK1/2-inactivating mutations were noted in tumor biopsies of 1 of 23 patients with melanoma and in 1 of 16 patients with mismatch repair-deficient colon cancer treated with PD-1 blockade. Both cases had a high mutational load but did not respond to anti-PD-1 therapy. Two out of 48 human melanoma cell lines had JAK1/2 mutations, which led to a lack of PD-L1 expression upon interferon gamma exposure mediated by an inability to signal through the interferon gamma receptor pathway. JAK1/2 loss-of-function alterations in The Cancer Genome Atlas confer adverse outcomes in patients. We propose that JAK1/2 loss-of-function mutations are a genetic mechanism of lack of reactive PD-L1 expression and response to interferon gamma, leading to primary resistance to PD-1 blockade therapy. SIGNIFICANCE: A key functional result from somatic JAK1/2 mutations in a cancer cell is the inability to respond to interferon gamma by expressing PD-L1 and many other interferon-stimulated genes. These mutations result in a genetic mechanism for the absence of reactive PD-L1 expression, and patients harboring such tumors would be unlikely to respond to PD-1 blockade therapy. Cancer Discov; 7(2); 188-201. ©2016 AACR.See related commentary by Marabelle et al., p. 128This article is highlighted in the In This Issue feature, p. 115.


Subject(s)
Drug Resistance, Neoplasm , Janus Kinase 1/genetics , Janus Kinase 2/genetics , Mutation , Neoplasms/genetics , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Antibodies, Monoclonal, Humanized/pharmacology , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Humans , Interferon-gamma/pharmacology , Melanoma/drug therapy , Melanoma/genetics , Neoplasms/drug therapy , Signal Transduction/drug effects
8.
N Engl J Med ; 375(9): 819-29, 2016 Sep 01.
Article in English | MEDLINE | ID: mdl-27433843

ABSTRACT

BACKGROUND: Approximately 75% of objective responses to anti-programmed death 1 (PD-1) therapy in patients with melanoma are durable, lasting for years, but delayed relapses have been noted long after initial objective tumor regression despite continuous therapy. Mechanisms of immune escape in this context are unknown. METHODS: We analyzed biopsy samples from paired baseline and relapsing lesions in four patients with metastatic melanoma who had had an initial objective tumor regression in response to anti-PD-1 therapy (pembrolizumab) followed by disease progression months to years later. RESULTS: Whole-exome sequencing detected clonal selection and outgrowth of the acquired resistant tumors and, in two of the four patients, revealed resistance-associated loss-of-function mutations in the genes encoding interferon-receptor-associated Janus kinase 1 (JAK1) or Janus kinase 2 (JAK2), concurrent with deletion of the wild-type allele. A truncating mutation in the gene encoding the antigen-presenting protein beta-2-microglobulin (B2M) was identified in a third patient. JAK1 and JAK2 truncating mutations resulted in a lack of response to interferon gamma, including insensitivity to its antiproliferative effects on cancer cells. The B2M truncating mutation led to loss of surface expression of major histocompatibility complex class I. CONCLUSIONS: In this study, acquired resistance to PD-1 blockade immunotherapy in patients with melanoma was associated with defects in the pathways involved in interferon-receptor signaling and in antigen presentation. (Funded by the National Institutes of Health and others.).


Subject(s)
Drug Resistance, Neoplasm/genetics , Immunotherapy , Janus Kinase 1/genetics , Janus Kinase 2/genetics , Melanoma/genetics , Mutation , Programmed Cell Death 1 Receptor/antagonists & inhibitors , beta 2-Microglobulin/genetics , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/therapeutic use , Biopsy , Exome , Gene Expression Regulation, Neoplastic , Genes, MHC Class I , Humans , Interferon-gamma/therapeutic use , Melanoma/drug therapy , Melanoma/secondary , Programmed Cell Death 1 Receptor/metabolism , Recurrence , Sequence Analysis, DNA , Signal Transduction
9.
J Immunother Cancer ; 4: 15, 2016.
Article in English | MEDLINE | ID: mdl-26981245

ABSTRACT

The efficacy of PD-1/PD-L1 targeted therapies in addition to anti-CTLA-4 solidifies immunotherapy as a modality to add to the anticancer arsenal. Despite raising the bar of clinical efficacy, immunologically targeted agents raise new challenges to conventional drug development paradigms by highlighting the limited relevance of assessing standard pharmacokinetics (PK) and pharmacodynamics (PD). Specifically, systemic and intratumoral immune effects have not consistently correlated with standard relationships between systemic dose, toxicity, and efficacy for cytotoxic therapies. Hence, PK and PD paradigms remain inadequate to guide the selection of doses and schedules, both starting and recommended Phase 2 for immunotherapies. The promise of harnessing the immune response against cancer must also be considered in light of unique and potentially serious toxicities. Refining immune endpoints to better inform clinical trial design represents a high priority challenge. The Cancer Immunotherapy Trials Network investigators review the immunodynamic effects of specific classes of immunotherapeutic agents to focus immune assessment modalities and sites, both systemic and importantly intratumoral, which are critical to the success of the rapidly growing field of immuno-oncology.

10.
Cancer Immunol Res ; 4(3): 194-203, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26787823

ABSTRACT

Tumor responses to programmed cell death protein 1 (PD-1) blockade therapy are mediated by T cells, which we characterized in 102 tumor biopsies obtained from 53 patients treated with pembrolizumab, an antibody to PD-1. Biopsies were dissociated, and single-cell infiltrates were analyzed by multicolor flow cytometry using two computational approaches to resolve the leukocyte phenotypes at the single-cell level. There was a statistically significant increase in the frequency of T cells in patients who responded to therapy. The frequency of intratumoral B cells and monocytic myeloid-derived suppressor cells significantly increased in patients' biopsies taken on treatment. The percentage of cells with a regulatory T-cell phenotype, monocytes, and natural killer cells did not change while on PD-1 blockade therapy. CD8(+) memory T cells were the most prominent phenotype that expanded intratumorally on therapy. However, the frequency of CD4(+) effector memory T cells significantly decreased on treatment, whereas CD4(+) effector T cells significantly increased in nonresponding tumors on therapy. In peripheral blood, an unusual population of blood cells expressing CD56 was detected in two patients with regressing melanoma. In conclusion, PD-1 blockade increases the frequency of T cells, B cells, and myeloid-derived suppressor cells in tumors, with the CD8(+) effector memory T-cell subset being the major T-cell phenotype expanded in patients with a response to therapy.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/therapeutic use , Melanoma/drug therapy , Skin Neoplasms/drug therapy , T-Lymphocyte Subsets/immunology , Aged , Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/pharmacology , Female , Humans , Male , Melanoma/immunology , Melanoma/secondary , Middle Aged , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Treatment Outcome
11.
Nature ; 515(7528): 568-71, 2014 Nov 27.
Article in English | MEDLINE | ID: mdl-25428505

ABSTRACT

Therapies that target the programmed death-1 (PD-1) receptor have shown unprecedented rates of durable clinical responses in patients with various cancer types. One mechanism by which cancer tissues limit the host immune response is via upregulation of PD-1 ligand (PD-L1) and its ligation to PD-1 on antigen-specific CD8(+) T cells (termed adaptive immune resistance). Here we show that pre-existing CD8(+) T cells distinctly located at the invasive tumour margin are associated with expression of the PD-1/PD-L1 immune inhibitory axis and may predict response to therapy. We analysed samples from 46 patients with metastatic melanoma obtained before and during anti-PD-1 therapy (pembrolizumab) using quantitative immunohistochemistry, quantitative multiplex immunofluorescence, and next-generation sequencing for T-cell antigen receptors (TCRs). In serially sampled tumours, patients responding to treatment showed proliferation of intratumoral CD8(+) T cells that directly correlated with radiographic reduction in tumour size. Pre-treatment samples obtained from responding patients showed higher numbers of CD8-, PD-1- and PD-L1-expressing cells at the invasive tumour margin and inside tumours, with close proximity between PD-1 and PD-L1, and a more clonal TCR repertoire. Using multivariate analysis, we established a predictive model based on CD8 expression at the invasive margin and validated the model in an independent cohort of 15 patients. Our findings indicate that tumour regression after therapeutic PD-1 blockade requires pre-existing CD8(+) T cells that are negatively regulated by PD-1/PD-L1-mediated adaptive immune resistance.


Subject(s)
Adaptive Immunity/immunology , CD8-Positive T-Lymphocytes/immunology , Immunotherapy , Melanoma/therapy , Models, Biological , Aged , Aged, 80 and over , Biomarkers , CD8-Positive T-Lymphocytes/cytology , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Melanoma/diagnosis , Melanoma/immunology , Melanoma/pathology , Middle Aged , Multivariate Analysis , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Treatment Outcome
12.
Oncoimmunology ; 3: e29244, 2014.
Article in English | MEDLINE | ID: mdl-25083336

ABSTRACT

Targeting immune inhibitory receptors has brought excitement, innovation and hope to cancer patients. Our recent work revealed the immunological effects of blocking the CTLA4 and PD-1 immune checkpoints on T cell receptor usage among peripheral blood cells, and further uncovers how the expansion of the T cell repertoire matches the immunotoxicity profile of the therapy.

13.
Clin Cancer Res ; 20(19): 4982-4, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-24970841

ABSTRACT

It is conceivable that, in the near future, an assay that defines the likelihood of a patient with advanced cancer to respond to immunotherapy based on PD1/L1 blockade will be the initial decision point to select the treatment of patients with any cancer type.


Subject(s)
Neoplasms/drug therapy , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , Humans , Molecular Targeted Therapy , Neoplasms/immunology , Neoplasms/metabolism , Patient Selection , Programmed Cell Death 1 Receptor/antagonists & inhibitors
14.
N Engl J Med ; 369(2): 134-44, 2013 Jul 11.
Article in English | MEDLINE | ID: mdl-23724846

ABSTRACT

BACKGROUND: The programmed death 1 (PD-1) receptor is a negative regulator of T-cell effector mechanisms that limits immune responses against cancer. We tested the anti-PD-1 antibody lambrolizumab (previously known as MK-3475) in patients with advanced melanoma. METHODS: We administered lambrolizumab intravenously at a dose of 10 mg per kilogram of body weight every 2 or 3 weeks or 2 mg per kilogram every 3 weeks in patients with advanced melanoma, both those who had received prior treatment with the immune checkpoint inhibitor ipilimumab and those who had not. Tumor responses were assessed every 12 weeks. RESULTS: A total of 135 patients with advanced melanoma were treated. Common adverse events attributed to treatment were fatigue, rash, pruritus, and diarrhea; most of the adverse events were low grade. The confirmed response rate across all dose cohorts, evaluated by central radiologic review according to the Response Evaluation Criteria in Solid Tumors (RECIST), version 1.1, was 38% (95% confidence interval [CI], 25 to 44), with the highest confirmed response rate observed in the cohort that received 10 mg per kilogram every 2 weeks (52%; 95% CI, 38 to 66). The response rate did not differ significantly between patients who had received prior ipilimumab treatment and those who had not (confirmed response rate, 38% [95% CI, 23 to 55] and 37% [95% CI, 26 to 49], respectively). Responses were durable in the majority of patients (median follow-up, 11 months among patients who had a response); 81% of the patients who had a response (42 of 52) were still receiving treatment at the time of analysis in March 2013. The overall median progression-free survival among the 135 patients was longer than 7 months. CONCLUSIONS: In patients with advanced melanoma, including those who had had disease progression while they had been receiving ipilimumab, treatment with lambrolizumab resulted in a high rate of sustained tumor regression, with mainly grade 1 or 2 toxic effects. (Funded by Merck Sharp and Dohme; ClinicalTrials.gov number, NCT01295827.).


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antineoplastic Agents/administration & dosage , Melanoma/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Skin Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Brain Neoplasms/secondary , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Humans , Male , Melanoma/secondary , Middle Aged , Skin Neoplasms/pathology
16.
Cancer Res ; 72(16): 3928-37, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22693252

ABSTRACT

Combining immunotherapy with targeted therapy blocking oncogenic BRAFV600 may result in improved treatments for advanced melanoma. In this study, we developed a BRAFV600E-driven murine model of melanoma, SM1, which is syngeneic to fully immunocompetent mice. SM1 cells exposed to the BRAF inhibitor vemurafenib (PLX4032) showed partial in vitro and in vivo sensitivity resulting from the inhibition of MAPK pathway signaling. Combined treatment of vemurafenib plus adoptive cell transfer therapy with lymphocytes genetically modified with a T-cell receptor (TCR) recognizing chicken ovalbumin (OVA) expressed by SM1-OVA tumors or pmel-1 TCR transgenic lymphocytes recognizing gp100 endogenously expressed by SM1 resulted in superior antitumor responses compared with either therapy alone. T-cell analysis showed that vemurafenib did not significantly alter the expansion, distribution, or tumor accumulation of the adoptively transferred cells. However, vemurafenib paradoxically increased mitogen-activated protein kinase (MAPK) signaling, in vivo cytotoxic activity, and intratumoral cytokine secretion by adoptively transferred cells. Taken together, our findings, derived from 2 independent models combining BRAF-targeted therapy with immunotherapy, support the testing of this therapeutic combination in patients with BRAFV600 mutant metastatic melanoma.


Subject(s)
Immunotherapy, Adoptive/methods , Indoles/pharmacology , Melanoma/therapy , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Sulfonamides/pharmacology , Animals , Combined Modality Therapy , Female , Humans , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Male , Melanoma/enzymology , Melanoma/immunology , Melanoma, Experimental/enzymology , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/genetics , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Tumor Cells, Cultured , Vemurafenib
17.
J Am Acad Dermatol ; 65(4): 722-725.e2, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21658797

ABSTRACT

BACKGROUND: Dermatologists at the University of California, San Francisco recently reported two patients in the online Journal of the American Academy of Dermatology with purpura presumably induced by levamisole in contaminated cocaine. Levamisole-induced vasculitis and neutropenia has been reported elsewhere in the United States and Canada. Up to 70% of cocaine in the United States could be contaminated. OBJECTIVE: We sought to describe similar cases of vasculitis associated with cocaine use. METHODS: This is a retrospective case series. RESULTS: We report 6 remarkably similar patients seen over just the past few months with retiform purpura on the body and tender purpuric eruptions, necrosis, and eschars of the ears after cocaine use in New York and California. All of these patients had positive perinuclear antineutrophil cytoplasmic antibody values and 3 of the 6 also had an associated neutropenia. Direct immunofluorescence studies suggested an immune complex-mediated vasculitis. LIMITATIONS: This case series is descriptive in nature and, because testing is not easily performed, we did not test for levamisole in the serum or blood to prove this is the causative agent. CONCLUSION: It appears the use of cocaine is associated with the peculiar clinical findings of ear purpura, retiform purpura of the trunk, and neutropenia. We believe this case series may represent the tip of the iceberg as a looming public health problem caused by levamisole. Although the direct causal relationship may be difficult to establish, the astute dermatologist or primary care physician should be able to recognize the characteristic skin lesions and should be wary of the potential development of agranulocytosis.


Subject(s)
Cocaine/adverse effects , Ear, External/drug effects , Levamisole/adverse effects , Neutropenia/chemically induced , Purpura/chemically induced , Vasculitis/chemically induced , Adult , Agranulocytosis/chemically induced , Cocaine-Related Disorders/complications , Drug Contamination , Female , Humans , Male , Middle Aged , Public Health
18.
J Immunother ; 33(8): 759-68, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20842061

ABSTRACT

Optimized conditions for the ex vivo activation, genetic manipulation, and expansion of human lymphocytes for adoptive cell therapy may lead to protocols that maximize their in vivo function. We analyzed the effects of 4 clinical grade activation and expansion protocols over 3 weeks on cell proliferative rate, immunophenotype, cell metabolism, and transduction efficiency of human peripheral blood mononuclear cells (PBMCs). Peak lentiviral transduction efficiency was early (days 2 to 4), at a time when cells showed a larger size, maximal uptake of metabolic substrates, and the highest level of proximal T-cell receptor signaling engagement. Anti-CD2/3/28 activation beads induced greater proliferation rate and skewed PBMCs early on to a CD4 phenotype when compared with the cells cultured in OKT3. Multicolor surface phenotyping demonstrated that changes in T-cell surface markers that define T-cell functional phenotypes were dependent on the time spent in culture as opposed to the particular activation protocol. In conclusion, ex vivo activation of human PBMCs for adoptive cell therapy demonstrate defined immunophenotypic and functional signatures over time, with cells early on showing larger sizes, higher transduction efficiency, maximal metabolic activity, and zeta-chain-associated protein-70 activation.


Subject(s)
Antineoplastic Protocols , Cell Culture Techniques , Genetic Therapy , Immunotherapy, Adoptive , T-Lymphocytes/metabolism , Antigens, CD/biosynthesis , Antigens, CD/immunology , Cell Proliferation , Cell Separation , Cells, Cultured , Flow Cytometry , Humans , Immunophenotyping , Lentivirus/genetics , Lymphocyte Activation , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/pathology , T-Lymphocytes/virology , Transduction, Genetic , ZAP-70 Protein-Tyrosine Kinase/metabolism
19.
J Nucl Med ; 49(6): 865-8, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18511842

ABSTRACT

Immune system activation can be elicited in viral infections, active immunization, or cancer immunotherapy, leading to the final common phenotype of increased glycolytic use by immune cells and subsequent detection by 18F-FDG PET. Because 18F-FDG is also used in baseline staging PET/CT scans and in tumor response assessment, physicians are faced with a unique challenge when evaluating tumor response in patients receiving cancer immunotherapy. The burgeoning field of cancer immunotherapy and the paucity of PET probes that can reliably differentiate activated immune cells from metabolically active cancer cells underscore the pressing need to identify and develop additional molecular imaging strategies. In an effort to address this concern, investigators have taken several molecular imaging approaches for cancer immunotherapy. Direct ex vivo labeling of T lymphocytes with radioactive probes before reinfusion represents the earliest attempts but has proven to be clinically limited because of significant PET probe dilution from proliferation of activated immune cells. Another approach is the indirect in vivo labeling of immune cells via PET reporter gene expression and involves the ex vivo genetic engineering of T lymphocytes with a reporter gene, reinfusion into the host, and the subsequent use of a PET probe specific for the reporter gene. The most recent approach involves the direct in vivo labeling of immune cells by targeting endogenous immune cell biochemical pathways that are differentially expressed during activation. In conclusion, these novel PET-based imaging approaches have demonstrated promise toward the goal of in vivo, noninvasive immune monitoring strategies for evaluating cancer immunotherapy.


Subject(s)
Biomedical Research/trends , Immunotherapy/trends , Molecular Probe Techniques/trends , Neoplasms/diagnostic imaging , Neoplasms/therapy , Positron-Emission Tomography/trends , Humans
20.
Cancer Biol Ther ; 6(3): 447-53, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17387267

ABSTRACT

Molecular imaging techniques can detect and monitor characteristics of the tumor microenvironment, such as angiogenesis, hypoxia, metabolism, and apoptosis that may better correlate with response to cancer therapy and may provide information in real-time. We investigated the use of a novel, spatially discrete, hyperspectral, multi-fiber optical system to characterize selected regions of skin in living mice. We determined the reproducibility and robustness of the spectral signatures derived from comparable regions of interest. Additionally, we characterized spectral differences in vascular and non-vascular fields to determine their potential use in monitoring angiogenesis. The macroscopic Prism and Reflectance Imaging Spectroscopy System (MACRO-PARISS) was calibrated against a National Institute for Standards and Technology (NIST)-certified lamp, allowing for reproducible spectra with any instrument similarly calibrated. Spectra were classified using a linearity-independent algorithm over a wavelength range of 450-920 nm. Classified spectra were integrated into a spectral library and subsequent acquisitions were correlated with the library set to a minimum correlation coefficient (MCC) of 99%. The results indicated that similar regions of interest with respect to vascularity consistently generated a unique spectral signature. As the field of view (FOV) moved from vascular to non-vascular areas, the acquired spectra changed in a step-wise and predictable fashion. Additionally, vascular fields that were deprived of their blood supply subsequently generated a non-vascular spectral signature. This work has implications for the monitoring of various physiologic or pathological processes including tumor angiogenesis and the therapeutic effects of anti-vascular agents.


Subject(s)
Fiber Optic Technology/instrumentation , Monitoring, Physiologic/instrumentation , Neovascularization, Physiologic , Skin/blood supply , Spectrum Analysis/methods , Animals , Mice , Mice, SCID , Reproducibility of Results
SELECTION OF CITATIONS
SEARCH DETAIL
...