2601
|
Berner AM, Sharma A, Agarwal S, Al-Sam S, Nathan P. Fatal autoimmune myocarditis with anti-PD-L1 and tyrosine kinase inhibitor therapy for renal cell cancer. Eur J Cancer 2018; 101:287-290. [PMID: 30017380 DOI: 10.1016/j.ejca.2018.06.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Revised: 06/13/2018] [Accepted: 06/19/2018] [Indexed: 11/18/2022]
Affiliation(s)
- A M Berner
- Department of Oncology, Mount Vernon Cancer Centre, Northwood HA6 2RN, UK.
| | - A Sharma
- Department of Oncology, Mount Vernon Cancer Centre, Northwood HA6 2RN, UK
| | - S Agarwal
- Department of Histopathology, East and North Hertfordshire NHS Trust, Coreys Mill Lane, Stevenage, Hertfordshire SG1 4AB, UK
| | - S Al-Sam
- Princess Alexandra Hospital, Hamstel Road, Harlow, Essex CM20 1QX, UK
| | - P Nathan
- Department of Oncology, Mount Vernon Cancer Centre, Northwood HA6 2RN, UK
| |
Collapse
|
2602
|
Abstract
INTRODUCTION In non-small cell lung cancer (NSCLC), immunotherapy is one of today's most important and ground-breaking systemic treatments, mainly represented by antibodies against cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed death protein 1 or ligand 1 (PD-1/PD-L1). Durvalumab (MEDI4736) is a high-affinity human IgG1 monoclonal antibody that binds to PD-1 and CD80, blocking PD-L1, but not PD-L2. Areas covered: In advanced NSCLC patients, durvalumab has demonstrated activity and acceptable tolerability, particularly with ≥25% PD-L1 tumor expression in the EGFR and ALK wild-type population. However, preliminary data have shown lower efficacy in EGFR mutant and ALK-positive patients. The results from the recent PACIFIC study in locally advanced patients have placed durvalumab as standard of care in consolidation after chemoradiation, leading to Food and Drug Administration (FDA) approval. Expert commentary: Early data suggest promising activity for durvalumab with the CTLA-4 inhibitor tremelimumab, regardless of PD-L1 expression, and potentially in combination with other drugs such as platinum-doublet chemotherapy. However, treatment-related toxicity associated with the combinations is an important aspect of the benefit-risk evaluation in the decision-making process. Results of ongoing phase III trials will provide illuminating data to confirm the place of durvalumab in the management of NSCLC patients.
Collapse
Affiliation(s)
- Laura Mezquita
- a Medical Oncology Department , Gustave Roussy , Villejuif , France
| | - David Planchard
- a Medical Oncology Department , Gustave Roussy , Villejuif , France
| |
Collapse
|
2603
|
Amini-Adle M, Piperno M, Tordo J, Thomas L, Dalle S, Dubois V, Marabelle A. Remitting Seronegative Symmetric Synovitis With Pitting Edema Associated With Partial Melanoma Response Under Anti-CTLA-4 and Anti-Programmed Death 1 Combination Treatment. Arthritis Rheumatol 2018; 70:1358. [DOI: 10.1002/art.40506] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
| | | | | | - Luc Thomas
- Hospices Civils de Lyon; Pierre Bénite France
| | | | - Valérie Dubois
- Etablissement Français du Sang Auvergne Rhone Alpes; Décines France
| | | |
Collapse
|
2604
|
Denaro N, Merlano MC. Immunotherapy in Head and Neck Squamous Cell Cancer. Clin Exp Otorhinolaryngol 2018; 11:217-223. [PMID: 29973040 PMCID: PMC6222190 DOI: 10.21053/ceo.2018.00150] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 03/26/2018] [Indexed: 01/07/2023] Open
Abstract
Prognosis in relapsed metastatic head and neck squamous cell cancer (RM-HNSCC) is dismal. Platinum based chemotherapy in combination with Cetuximab is used in first-line setting, while no further validated options are available at progression. Immunotherapy has produced durable clinical benefit in some patients with RM-HNSCC although the premises are several patients are nonresponders. Studies are ongoing to determine predictive factors and the ideal setting/combination of novel immunotherapies. In this paper, we discuss the past and present of immunotherapy in head and neck cancer and provide an up-to-date information regarding the potential ways to improve immunotherapy outcomes in HNSCC.
Collapse
Affiliation(s)
- Nerina Denaro
- Department of Oncology, ASO Santa Croce e Carle, Cuneo, Italy
| | | |
Collapse
|
2605
|
Karamouzis MV, Papavassiliou AG. Combination of checkpoint inhibitors with other agents as a strategy to improve anti-cancer effect - a glimpse to the future. Expert Opin Investig Drugs 2018; 27:569-572. [PMID: 29958097 DOI: 10.1080/13543784.2018.1494724] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
In the last years, a remarkable progress has been made in the clinical application of novel immunotherapy agents, the so called 'checkpoint inhibitors,' that has revolutionized the treatment of many malignant tumors. Their design has been based on the immune-mediated mechanisms of antitumor activity circle, such as antigen release and presentation, activation and trafficking of T-cells into tumors, depletion of immunosuppression, and immunogenic cell death. Various combinations of checkpoint inhibitors are being designed and/or tested, such as double checkpoint blockade, combination with chemotherapy, radiotherapy, molecularly targeted agents, and other immune-directed strategies.
Collapse
Affiliation(s)
- Michalis V Karamouzis
- a Molecular Oncology Unit, Department of Biological Chemistry , Medical School, National and Kapodistrian University of Athens , Athens , Greece
| | - Athanasios G Papavassiliou
- a Molecular Oncology Unit, Department of Biological Chemistry , Medical School, National and Kapodistrian University of Athens , Athens , Greece
| |
Collapse
|
2606
|
Pickwell-Smith BA, So ACP, Board RE. Managing side effects of cancer immunotherapy for the acute physician. Br J Hosp Med (Lond) 2018; 79:372-377. [DOI: 10.12968/hmed.2018.79.7.372] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Benjamin A Pickwell-Smith
- Specialist Registrar in Medical Oncology, Department of Oncology, Lancashire Teaching Hospitals NHS Trust, Fulwood, Preston
| | - Alfred CP So
- Medical Student, Faculty of Biology, Medicine and Health, University of Manchester, Manchester
| | - Ruth E Board
- Consultant Medical Oncologist, Department of Oncology, Lancashire Teaching Hospitals NHS Trust, Fulwood, Preston PR2 9HT and Honorary Senior Lecturer Faculty of Biology, Medicine and Health, University of Manchester, Manchester
| |
Collapse
|
2607
|
Induction of oligoclonal CD8 T cell responses against pulmonary metastatic cancer by a phospholipid-conjugated TLR7 agonist. Proc Natl Acad Sci U S A 2018; 115:E6836-E6844. [PMID: 29967183 DOI: 10.1073/pnas.1803281115] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Recent advances in cancer immunotherapy have improved patient survival. However, only a minority of patients with pulmonary metastatic disease respond to treatment with checkpoint inhibitors. As an alternate approach, we have tested the ability of systemically administered 1V270, a toll-like receptor 7 (TLR7) agonist conjugated to a phospholipid, to inhibit lung metastases in two variant murine 4T1 breast cancer models, as well as in B16 melanoma, and Lewis lung carcinoma models. In the 4T1 breast cancer models, 1V270 therapy inhibited lung metastases if given up to a week after primary tumor initiation. The treatment protocol was facilitated by the minimal toxic effects exerted by the phospholipid TLR7 agonist compared with the unconjugated agonist. 1V270 exhibited a wide therapeutic window and minimal off-target receptor binding. The 1V270 therapy inhibited colonization by tumor cells in the lungs in an NK cell dependent manner. Additional experiments revealed that single administration of 1V270 led to tumor-specific CD8+ cell-dependent adaptive immune responses that suppressed late-stage metastatic tumor growth in the lungs. T cell receptor (TCR) repertoire analyses showed that 1V270 therapy induced oligoclonal T cells in the lungs and mediastinal lymph nodes. Different animals displayed commonly shared TCR clones following 1V270 therapy. Intranasal administration of 1V270 also suppressed lung metastasis and induced tumor-specific adaptive immune responses. These results indicate that systemic 1V270 therapy can induce tumor-specific cytotoxic T cell responses to pulmonary metastatic cancers and that TCR repertoire analyses can be used to monitor, and to predict, the response to therapy.
Collapse
|
2608
|
Immune checkpoint blockade therapy for cancer: An overview of FDA-approved immune checkpoint inhibitors. Int Immunopharmacol 2018; 62:29-39. [PMID: 29990692 DOI: 10.1016/j.intimp.2018.06.001] [Citation(s) in RCA: 854] [Impact Index Per Article: 122.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 05/31/2018] [Accepted: 06/01/2018] [Indexed: 12/11/2022]
Abstract
Although T lymphocytes have long been appreciated for their role in the immunosurveillance of cancer, it has been the realization that cancer cells may ultimately escape a response from tumor-reactive T cells that has ignited efforts to enhance the efficacy of anti-tumor immune responses. Recent advances in our understanding of T cell immunobiology have been particularly instrumental in informing therapeutic strategies to overcome mechanisms of tumor immune escape, and immune checkpoint blockade has emerged as one of the most promising therapeutic options for patients in the history of cancer treatment. Designed to interfere with inhibitory pathways that naturally constrain T cell reactivity, immune checkpoint blockade releases inherent limits on the activation and maintenance of T cell effector function. In the context of cancer, where negative T cell regulatory pathways are often overactive, immune checkpoint blockade has proven to be an effective strategy for enhancing the effector activity and clinical impact of anti-tumor T cells. Checkpoint inhibitors targeting CTLA-4, PD-1, and PD-L1 have yielded unprecedented and durable responses in a significant percentage of cancer patients in recent years, leading to U.S. FDA approval of six checkpoint inhibitors for numerous cancer indications since 2011. In this review, we highlight the clinical success of these FDA-approved immune checkpoint inhibitors and discuss current challenges and future strategies that must be considered going forward to maximize the efficacy of immune checkpoint blockade therapy for cancer.
Collapse
|
2609
|
Cohen NA, Strong VE, Janjigian YY. Checkpoint blockade in esophagogastric cancer. J Surg Oncol 2018; 118:77-85. [PMID: 29878357 PMCID: PMC7891842 DOI: 10.1002/jso.25116] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 04/25/2018] [Indexed: 12/21/2022]
Abstract
There are few effective treatment options for metastatic esophagogastric adenocarcinomas after progression on second-line chemotherapy. Immune checkpoint blockade therapy is a promising treatment strategy for selected advanced esophagogastric cancer, and the PD-1 inhibitor pembrolizumab has recently been approved for metastatic or recurrent gastric or gastroesophageal junction cancer that has progressed beyond second-line systemic therapy. We review the current data supporting immune checkpoint blockade therapy in advanced esophagogastric adenocarcinoma.
Collapse
Affiliation(s)
- Noah A. Cohen
- Department of Surgery, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, New York
| | - Vivian E. Strong
- Department of Surgery, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, New York
| | - Yelena Y. Janjigian
- Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, New York
| |
Collapse
|
2610
|
Tumor-infiltrating lymphocytes and ductal carcinoma in situ of the breast: friends or foes? Mod Pathol 2018; 31:1012-1025. [PMID: 29463884 DOI: 10.1038/s41379-018-0030-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 01/05/2018] [Accepted: 01/07/2018] [Indexed: 12/11/2022]
Abstract
In the past three decades, the detection rate of ductal carcinoma in situ of the breast has dramatically increased due to breast screening programs. As a consequence, about 20% of all breast cancer cases are detected in this early in situ stage. Some ductal carcinoma in situ cases will progress to invasive breast cancer, while other cases are likely to have an indolent biological behavior. The presence of tumor-infiltrating lymphocytes is seen as a promising prognostic and predictive marker in invasive breast cancer, mainly in HER2-positive and triple-negative subtypes. Here, we summarize the current understanding regarding immune infiltrates in invasive breast cancer and highlight recent observations regarding the presence and potential clinical significance of such immune infiltrates in patients with ductal carcinoma in situ. The presence of tumor-infiltrating lymphocytes, their numbers, composition, and potential relationship with genomic status will be discussed. Finally, we propose that a combination of genetic and immune markers may better stratify ductal carcinoma in situ subtypes with respect to tumor evolution.
Collapse
|
2611
|
Twumasi-Boateng K, Pettigrew JL, Kwok YYE, Bell JC, Nelson BH. Oncolytic viruses as engineering platforms for combination immunotherapy. Nat Rev Cancer 2018; 18:419-432. [PMID: 29695749 DOI: 10.1038/s41568-018-0009-4] [Citation(s) in RCA: 283] [Impact Index Per Article: 40.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
To effectively build on the recent successes of immune checkpoint blockade, adoptive T cell therapy and cancer vaccines, it is critical to rationally design combination strategies that will increase and extend efficacy to a larger proportion of patients. For example, the combination of anti-cytotoxic T lymphocyte-associated antigen 4 (CTLA4) and anti-programmed cell death protein 1 (PD1) immune checkpoint inhibitors essentially doubles the response rate in certain patients with metastatic melanoma. However, given the heterogeneity of cancer, it seems likely that even more complex combinations of immunomodulatory agents may be required to obtain consistent, durable therapeutic responses against a broad spectrum of cancers. This carries serious implications in terms of toxicities for patients, feasibility for care providers and costs for health-care systems. A compelling solution is offered by oncolytic viruses (OVs), which can be engineered to selectively replicate within and destroy tumour tissue while simultaneously augmenting antitumour immunity. In this Opinion article, we argue that the future of immunotherapy will include OVs that function as multiplexed immune-modulating platforms expressing factors such as immune checkpoint inhibitors, tumour antigens, cytokines and T cell engagers. We illustrate this concept by following the trials and tribulations of tumour-reactive T cells from their initial priming through to the execution of cytotoxic effector function in the tumour bed. We highlight the myriad opportunities for OVs to help overcome critical barriers in the T cell journey, leading to new synergistic mechanisms in the battle against cancer.
Collapse
Affiliation(s)
- Kwame Twumasi-Boateng
- Trev and Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, British Columbia, Canada
| | - Jessica L Pettigrew
- Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Y Y Eunice Kwok
- Trev and Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, British Columbia, Canada
| | - John C Bell
- Center for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada.
| | - Brad H Nelson
- Trev and Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, British Columbia, Canada.
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia, Canada.
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
2612
|
Actualités autour des inhibiteurs de checkpoints immunitaires : enseignements issus du congrès ASCO 2017 et perspectives. Bull Cancer 2018; 105:686-695. [DOI: 10.1016/j.bulcan.2018.04.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 04/09/2018] [Accepted: 04/11/2018] [Indexed: 11/18/2022]
|
2613
|
Wagner NB, Forschner A, Leiter U, Garbe C, Eigentler TK. S100B and LDH as early prognostic markers for response and overall survival in melanoma patients treated with anti-PD-1 or combined anti-PD-1 plus anti-CTLA-4 antibodies. Br J Cancer 2018; 119:339-346. [PMID: 29950611 PMCID: PMC6070917 DOI: 10.1038/s41416-018-0167-x] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 06/07/2018] [Accepted: 06/07/2018] [Indexed: 01/19/2023] Open
Abstract
Background Immunotherapy with PD-1 antibodies has greatly increased prognosis of patients with advanced melanoma. Identifying biomarkers that predict overall survival (OS) and response to immunotherapy is important. Methods OS and best overall response according to RECIST version 1.1 were analysed, and S100B and lactate dehydrogenase (LDH) serum levels were assessed retrospectively in 152 patients treated with anti-PD-1, and in 86 patients treated with anti-PD-1 plus anti-CTLA-4 antibodies at University Hospital Tuebingen, Germany. Results In the pembrolizumab group, patients with elevated baseline S100B or LDH exhibited significantly impaired OS compared with patients with normal S100B (1-year OS: 51.1% vs 83.1%, log-rank P < .0001) and normal LDH (1-year OS: 44.4% vs 80.8%, P = .00022), respectively. LDH increases of >25% and S100B increases of >145% compared to baseline were significantly associated with impaired OS (both P < .0001). In patients treated with ipilimumab and nivolumab, baseline S100B and increasing S100B levels of >145% as well as baseline LDH were associated with impaired OS (P < .0001, P = .00060, and P = .0050, respectively), whereas increasing LDH of >25% was not (P = .64). Conclusions S100B could serve as a strong baseline marker for OS in melanoma patients receiving anti-PD-1 therapy. Rising S100B levels during the first weeks of therapy could help guide treatment decisions.
Collapse
Affiliation(s)
- Nikolaus B Wagner
- Department of Dermatology, University Hospital Tuebingen, Liebermeisterstrasse 25, 72076, Tuebingen, Germany.
| | - Andrea Forschner
- Department of Dermatology, University Hospital Tuebingen, Liebermeisterstrasse 25, 72076, Tuebingen, Germany
| | - Ulrike Leiter
- Department of Dermatology, University Hospital Tuebingen, Liebermeisterstrasse 25, 72076, Tuebingen, Germany
| | - Claus Garbe
- Department of Dermatology, University Hospital Tuebingen, Liebermeisterstrasse 25, 72076, Tuebingen, Germany
| | - Thomas K Eigentler
- Department of Dermatology, University Hospital Tuebingen, Liebermeisterstrasse 25, 72076, Tuebingen, Germany
| |
Collapse
|
2614
|
Berghoff AS, Preusser M. New developments in brain metastases. Ther Adv Neurol Disord 2018; 11:1756286418785502. [PMID: 30034538 PMCID: PMC6048670 DOI: 10.1177/1756286418785502] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 04/11/2018] [Indexed: 12/25/2022] Open
Abstract
Patients with brain metastases (BM) are a population of high clinical need for new therapeutic approaches due to, as yet, very impaired survival prognosis. However, only few clinical trials have specifically addressed this prognostically highly heterogeneous patient population. New developments in the treatment of BM patients aim to reduce the side effects of local therapies, for example, by redefining the indications for stereotactic radiosurgery and whole-brain radiotherapy (WBRT) or introducing new applications like hippocampal sparing WBRT. Furthermore, systemic therapies become a more important treatment approach in patients harboring targetable mutations, as recent BM-specific endpoints in several phase III trials have shown promising intracranial efficacy. In addition, immune-checkpoint inhibitors show promising intracranial efficacy, particularly in patients with melanoma and non-small lung cancer BM. Here, we provide a review on the recent new developments in the local and systemic therapy approaches in BM patients.
Collapse
Affiliation(s)
- Anna S. Berghoff
- Department of Medicine I, Medical University of
Vienna, Vienna, Austria Comprehensive Cancer Center, Medical University of
Vienna, Vienna, Austria
| | - Matthias Preusser
- Department of Medicine I and Comprehensive
Cancer Center CNS Unit (CCC-CNS), Medical University of Vienna, Waehringer
Guertel 18-20, 1090 Vienna, Austria
| |
Collapse
|
2615
|
Buder-Bakhaya K, Hassel JC. Biomarkers for Clinical Benefit of Immune Checkpoint Inhibitor Treatment-A Review From the Melanoma Perspective and Beyond. Front Immunol 2018; 9:1474. [PMID: 30002656 PMCID: PMC6031714 DOI: 10.3389/fimmu.2018.01474] [Citation(s) in RCA: 167] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 06/13/2018] [Indexed: 12/26/2022] Open
Abstract
Background Immune checkpoint inhibition (ICI) with anti-CTLA-4 and/or anti-PD-1 antibodies is standard treatment for metastatic melanoma. Anti-PD-1 (pembrolizumab, nivolumab) and anti-PD-L1 antibodies (atezolizumab, durvalumab, and avelumab) have been approved for treatment of several other advanced malignancies, including non-small-cell lung cancer (NSCLC); renal cell, and urothelial carcinoma; head and neck cancer; gastric, hepatocellular, and Merkel-cell carcinoma; and classical Hodgkin lymphoma. In some of these malignancies approval was based on the detection of biomarkers such as PD-L1 expression or high microsatellite instability. Methods We review the current status of prognostic and predictive biomarkers used in ICI for melanoma and other malignancies. We include clinical, tissue, blood, and stool biomarkers, as well as imaging biomarkers. Results Several biomarkers have been studied in ICI for metastatic melanoma. In clinical practice, pre-treatment tumor burden measured by means of imaging and serum lactate dehydrogenase level is already being used to estimate the likelihood of effective ICI treatment. In peripheral blood, the number of different immune cell types, such as lymphocytes, neutrophils, and eosinophils, as well as different soluble factors, have been correlated with clinical outcome. For intra-tumoral biomarkers, expression of the PD-1 ligand PD-L1 has been found to be of some predictive value for anti-PD-1-directed therapy for NSCLC and melanoma. A high mutational load, particularly when accompanied by neoantigens, seems to facilitate immune response and correlates with patient survival for all entities treated by use of ICI. Tumor microenvironment also seems to be of major importance. Interestingly, even the gut microbiome has been found to correlate with response to ICI, most likely through immuno-stimulatory effects of distinct bacteria. New imaging biomarkers, e.g., for PET, and magnetic resonance imaging are also being investigated, and results suggest they will make early prediction of patient response possible. Conclusion Several promising results are available regarding possible biomarkers for response to ICI, which need to be validated in large clinical trials. A better understanding of how ICI works will enable the development of biomarkers that can predict the response of individual patients.
Collapse
Affiliation(s)
- Kristina Buder-Bakhaya
- Section of Dermatooncology, Department of Dermatology and National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
| | - Jessica C Hassel
- Section of Dermatooncology, Department of Dermatology and National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
2616
|
Samra B, Tam E, Baseri B, Shapira I. Checkpoint inhibitors in head and neck cancer: current knowledge and perspectives. J Investig Med 2018; 66:1023-1030. [PMID: 29941547 DOI: 10.1136/jim-2018-000743] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/27/2018] [Indexed: 01/22/2023]
Abstract
The emergence of immunotherapy has provided significant clinical improvements in the treatment of metastatic solid tumors. Recurrent/metastatic head and neck squamous cell carcinoma (HNSCC) has dismal prognosis with median survival ranging between 6and12 months. Our aim is to review the current knowledge on the role of the immune system and immune checkpoint inhibitors in HNSCC. We will focus on the landmark trials that led to the regulatory approvals of pembrolizumab and nivolumab, and discuss a few promising contenders in clinical development and highlight the need to identify better biomarkers other than programmed death-ligand 1 to improve patient selection and help predict response.
Collapse
Affiliation(s)
- Bachar Samra
- Department of Hematology/Oncology, SUNY Downstate Medical Center, Brooklyn, New York, USA.,Department of Hematology/Oncology, SUNY Downstate Medical Center College of Medicine, Brooklyn, New York, USA
| | - Eric Tam
- Department of Hematology/Oncology, SUNY Downstate Medical Center, Brooklyn, New York, USA.,Department of Hematology/Oncology, SUNY Downstate Medical Center College of Medicine, Brooklyn, New York, USA
| | - Babak Baseri
- Department of Hematology/Oncology, SUNY Downstate Medical Center, Brooklyn, New York, USA.,Department of Hematology/Oncology, SUNY Downstate Medical Center College of Medicine, Brooklyn, New York, USA
| | - Iuliana Shapira
- Department of Hematology/Oncology, SUNY Downstate Medical Center College of Medicine, Brooklyn, New York, USA
| |
Collapse
|
2617
|
Kirchberger MC, Moreira A, Erdmann M, Schuler G, Heinzerling L. Real world experience in low-dose ipilimumab in combination with PD-1 blockade in advanced melanoma patients. Oncotarget 2018; 9:28903-28909. [PMID: 29988983 PMCID: PMC6034742 DOI: 10.18632/oncotarget.25627] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Accepted: 05/30/2018] [Indexed: 12/14/2022] Open
Abstract
Dual immune-checkpoint blockade with the anti-PD-1 antibody nivolumab (1 mg/kg) and standard-dose ipilimumab (3 mg/kg) is the mainstay of immunotherapy in advanced melanoma and it is approved since 2016. However, severe side effects (grade 3/4) occur in up to 60% of the patients. Recently, clinical trials have shown similar anti-tumor activity with a more favorable toxicity profile in patients treated with low-dose ipilimumab (1 mg/kg) and standard-dose pembrolizumab (2 mg/kg). In this study we report on the real-world experience of this dosing regime in advanced melanoma patients not eligible for clinical trials. A total of 33 patients with metastatic melanoma (24 with cutaneous and 9 with uveal melanoma) were assessed, retrospectively. Brain metastases were present in 33% of the patients and lactate dehydrogenase was elevated in 70%. Overall response rates were 38% and 0% in cutaneous melanoma and uveal melanoma respectively. Median overall survival was not reached in cutaneous melanoma and was 18 months in uveal melanoma. In 18% of the patients at least one treatment-related severe adverse event was observed. Our observation that the combination of standard dose pembrolizumab and low-dose ipilimumab has a favorable toxicity profile yet anti-tumor activity comparable to the approved standard-dose combination regime in advanced patients not suitable for enrollment in clinical trials is encouraging.
Collapse
Affiliation(s)
- Michael Constantin Kirchberger
- Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Ulmenweg 18, 91054 Erlangen, Germany
| | - Alvaro Moreira
- Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Ulmenweg 18, 91054 Erlangen, Germany
| | - Michael Erdmann
- Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Ulmenweg 18, 91054 Erlangen, Germany
| | - Gerold Schuler
- Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Ulmenweg 18, 91054 Erlangen, Germany
| | - Lucie Heinzerling
- Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Ulmenweg 18, 91054 Erlangen, Germany
| |
Collapse
|
2618
|
Sato Y, Fujimura T, Kambayashi Y, Tanita K, Tono H, Hashimoto A, Aiba S. Two cases of dabrafenib and trametinib therapy-failed advanced melanoma successfully controlled by nivolumab monotherapy. J Dermatol 2018; 45:1105-1108. [DOI: 10.1111/1346-8138.14508] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 05/14/2018] [Indexed: 01/26/2023]
Affiliation(s)
- Yota Sato
- Department of Dermatology; Tohoku University Graduate School of Medicine; Sendai Japan
| | - Taku Fujimura
- Department of Dermatology; Tohoku University Graduate School of Medicine; Sendai Japan
| | - Yumi Kambayashi
- Department of Dermatology; Tohoku University Graduate School of Medicine; Sendai Japan
| | - Kayo Tanita
- Department of Dermatology; Tohoku University Graduate School of Medicine; Sendai Japan
| | - Hisayuki Tono
- Department of Dermatology; Tohoku University Graduate School of Medicine; Sendai Japan
| | - Akira Hashimoto
- Department of Dermatology; Tohoku University Graduate School of Medicine; Sendai Japan
| | - Setsuya Aiba
- Department of Dermatology; Tohoku University Graduate School of Medicine; Sendai Japan
| |
Collapse
|
2619
|
Fumet JD, Isambert N, Hervieu A, Zanetta S, Guion JF, Hennequin A, Rederstorff E, Bertaut A, Ghiringhelli F. Phase Ib/II trial evaluating the safety, tolerability and immunological activity of durvalumab (MEDI4736) (anti-PD-L1) plus tremelimumab (anti-CTLA-4) combined with FOLFOX in patients with metastatic colorectal cancer. ESMO Open 2018; 3:e000375. [PMID: 29942666 PMCID: PMC6012564 DOI: 10.1136/esmoopen-2018-000375] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 04/25/2018] [Accepted: 04/27/2018] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND 5-Fluorouracil plus irinotecan or oxaliplatin alone or in association with target therapy are standard first-line therapy for metastatic colorectal cancer (mCRC). Checkpoint inhibitors targeting PD-1/PD-L1 demonstrated efficacy on mCRC with microsatellite instability but remain ineffective alone in microsatellite stable tumour. 5-Fluorouracil and oxaliplatin were known to present immunogenic properties. Durvalumab (D) is a human monoclonal antibody (mAb) that inhibits binding of programmed cell death ligand 1 (PD-L1) to its receptor. Tremelimumab (T) is a mAb directed against the cytotoxic T-lymphocyte-associated protein 4 (CTLA-4). This study is designed to evaluate whether the addition of PD-L1 and CTLA-4 inhibition to oxaliplatin, fluorouracil and leucovorin (FOLFOX) increases treatment efficacy. METHODS This phase II study (ClinicalTrials.gov NCT03202758) will assess the efficacy and safety of FOLFOX/D/T association in patients with mCRC (n=48). Good performance status patients (Eastern Cooperative Oncology Group <2) with untreated, RAS mutational status mCRC will be eligible. Prior adjuvant therapy is allowed provided recurrence is >6 months postcompletion. There is a safety lead in nine patients receiving FOLFOX/D/T. Assuming no safety concerns the study will go on to include 39 additional patients. Patients will receive folinic acid (400 mg/m²)/5-fluorouracil (400 mg/m² as bolus followed by 2400 mg/m2 as a 46-hour infusion)/oxaliplatin (85 mg/m2) every 14 days with D (750 mg) D1 every 14 days and T (75 mg) D1 every 28 days. After six cycles of FOLFOX only D/T will continue until disease progression, death, intolerable toxicity, or patient/investigator decision to stop. Primary endpoint is safety and efficacy according to progression-free survival (PFS); secondary endpoints include overall response rate and quality of life. Hypothesis is that a PFS of 50% at 6 months is insufficient and a PFS of 70.7% is expected (with α=10%, β=10%). Blood, plasma and tumour tissue will be collected and assessed for potential prognostic and predictive biomarkers.
Collapse
Affiliation(s)
- Jean-David Fumet
- Department of Medical Oncology, Center Georges Francois Leclerc, Dijon, France
- Research Platform in Biological Oncology, Georges Francois Leclerc Center, Dijon, France
- University of Burgundy-Franche Comté, Dijon, France
| | - Nicolas Isambert
- Department of Medical Oncology, Center Georges Francois Leclerc, Dijon, France
| | - Alice Hervieu
- Department of Medical Oncology, Center Georges Francois Leclerc, Dijon, France
| | - Sylvie Zanetta
- Department of Medical Oncology, Center Georges Francois Leclerc, Dijon, France
| | - Jean-Florian Guion
- Department of Medical Oncology, Center Georges Francois Leclerc, Dijon, France
| | - Audrey Hennequin
- Department of Medical Oncology, Center Georges Francois Leclerc, Dijon, France
| | - Emilie Rederstorff
- Department of Epidemiology and Biostatistics, Georges François Leclerc Center, Dijon, France
| | - Aurélie Bertaut
- Department of Epidemiology and Biostatistics, Georges François Leclerc Center, Dijon, France
| | - Francois Ghiringhelli
- Department of Medical Oncology, Center Georges Francois Leclerc, Dijon, France
- Research Platform in Biological Oncology, Georges Francois Leclerc Center, Dijon, France
- University of Burgundy-Franche Comté, Dijon, France
- INSERM UMR1231, Dijon, France
- GIMI Genetic and Immunology Medical Institute, Dijon, France
| |
Collapse
|
2620
|
Groisberg R, Subbiah V. Immunotherapy and next-generation sequencing guided therapy for precision oncology: What have we learnt and what does the future hold? EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2018; 3:205-213. [PMID: 31886407 PMCID: PMC6934166 DOI: 10.1080/23808993.2018.1480898] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
INTRODUCTION The rapid development of clinical next generation sequencing (NGS) and the contemporaneous availability of molecular targeted therapies ignited and fueled the field of precision oncology. More recently there has been an explosion of immunotherapeutic agents, specifically the checkpoint inhibitors: PD-1, PD-L1 and CTLA-4 antibodies. These new classes of agents have produced durable responses in a variety of tumor subtypes. AREAS COVERED In this review, the authors explore the role of NGS in identifying targets for molecular therapy. The authors also expand on the future uses of NGS in oncology including: prediction of checkpoint inhibitor response, quantification of tumor mutational burden, neoantigen calling using bioinformatics tools, and finally the personalization of cell transfer technologies and cancer vaccines. EXPERT COMMENTARY The near future will witness an increased understanding of the immune system and genomics in cancer. High throughput sequencing technology will expand in parallel with an ever-expanding array of novel therapies. Improved computational power coupled with bioinformatics algorithms will combine the fields of genomics and immunology. The emerging fields that stand to benefit from rapid translation of NGS technology include cancer vaccines and adoptive cell therapy, which will further refine precision oncology.
Collapse
Affiliation(s)
- Roman Groisberg
- Department of Investigational Cancer Therapeutics (Phase 1 Program), Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Vivek Subbiah
- Department of Investigational Cancer Therapeutics (Phase 1 Program), Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
2621
|
Zappasodi R, Budhu S, Hellmann MD, Postow MA, Senbabaoglu Y, Manne S, Gasmi B, Liu C, Zhong H, Li Y, Huang AC, Hirschhorn-Cymerman D, Panageas KS, Wherry EJ, Merghoub T, Wolchok JD. Non-conventional Inhibitory CD4 +Foxp3 -PD-1 hi T Cells as a Biomarker of Immune Checkpoint Blockade Activity. Cancer Cell 2018; 33:1017-1032.e7. [PMID: 29894689 PMCID: PMC6648657 DOI: 10.1016/j.ccell.2018.05.009] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 01/22/2018] [Accepted: 05/15/2018] [Indexed: 12/13/2022]
Abstract
A significant proportion of cancer patients do not respond to immune checkpoint blockade. To better understand the molecular mechanisms underlying these treatments, we explored the role of CD4+Foxp3- T cells expressing PD-1 (4PD1hi) and observed that 4PD1hi accumulate intratumorally as a function of tumor burden. Interestingly, CTLA-4 blockade promotes intratumoral and peripheral 4PD1hi increases in a dose-dependent manner, while combination with PD-1 blockade mitigates this effect and improves anti-tumor activity. We found that lack of effective 4PD1hi reduction after anti-PD-1 correlates with poor prognosis. Mechanistically, we provide evidence that mouse and human circulating and intra-tumor 4PD1hi inhibit T cell functions in a PD-1/PD-L1 dependent fashion and resemble follicular helper T cell (TFH)-like cells. Accordingly, anti-CTLA-4 activity is improved in TFH deficient mice.
Collapse
MESH Headings
- Animals
- Antibodies/pharmacology
- CD4-Positive T-Lymphocytes/drug effects
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- Cell Line, Tumor
- Forkhead Transcription Factors/genetics
- Forkhead Transcription Factors/immunology
- Forkhead Transcription Factors/metabolism
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Expression Regulation, Neoplastic/immunology
- Humans
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Neoplasms/drug therapy
- Neoplasms/genetics
- Neoplasms/immunology
- Programmed Cell Death 1 Receptor/genetics
- Programmed Cell Death 1 Receptor/immunology
- Programmed Cell Death 1 Receptor/metabolism
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
Collapse
Affiliation(s)
- Roberta Zappasodi
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Sadna Budhu
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Matthew D Hellmann
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Weill Cornell Medicine, New York, NY 10065, USA
| | - Michael A Postow
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Weill Cornell Medicine, New York, NY 10065, USA
| | - Yasin Senbabaoglu
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Sasikanth Manne
- Department of Microbiology and Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Billel Gasmi
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Cailian Liu
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Hong Zhong
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Yanyun Li
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Alexander C Huang
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Microbiology and Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Daniel Hirschhorn-Cymerman
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Katherine S Panageas
- Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - E John Wherry
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Microbiology and Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Taha Merghoub
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| | - Jedd D Wolchok
- Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Weill Cornell Medicine, New York, NY 10065, USA.
| |
Collapse
|
2622
|
Temel JS, Gainor JF, Sullivan RJ, Greer JA. Keeping Expectations in Check With Immune Checkpoint Inhibitors. J Clin Oncol 2018; 36:1654-1657. [DOI: 10.1200/jco.2017.76.2146] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Jennifer S. Temel
- Jennifer S. Temel, Justin F. Gainor, Ryan J. Sullivan, and Joseph A. Greer, Massachusetts General Hospital, Boston, MA
| | - Justin F. Gainor
- Jennifer S. Temel, Justin F. Gainor, Ryan J. Sullivan, and Joseph A. Greer, Massachusetts General Hospital, Boston, MA
| | - Ryan J. Sullivan
- Jennifer S. Temel, Justin F. Gainor, Ryan J. Sullivan, and Joseph A. Greer, Massachusetts General Hospital, Boston, MA
| | - Joseph A. Greer
- Jennifer S. Temel, Justin F. Gainor, Ryan J. Sullivan, and Joseph A. Greer, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
2623
|
Napolitano S, Brancaccio G, Argenziano G, Martinelli E, Morgillo F, Ciardiello F, Troiani T. It is finally time for adjuvant therapy in melanoma. Cancer Treat Rev 2018; 69:101-111. [PMID: 29957365 DOI: 10.1016/j.ctrv.2018.06.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 06/05/2018] [Accepted: 06/08/2018] [Indexed: 12/23/2022]
Abstract
Although melanoma is amenable to early detection, there has been no decline in the mortality rate of this disease and the prognosis of patients with high-risk primary melanoma or with macroscopic nodal involvement remains poor. The best option for patients with higher-risk melanoma is to receive effective adjuvant therapy in order to reduce their chances of recurrence. Multiple systemic therapeutic agents have been tested as adjuvant therapy for melanoma with durable benefits seen only with interferon- to date. More recently ipilimumab at the high dose of 10 mg/kg has shown a significant improvement in terms of Relapse free survival and Overall survival for stage III melanoma patients but at a significant cost in terms of immune-related toxicities. More recently, novel treatment options have emerged. The results from the latest trials with immunotherapy (PD-1 inhibitors) and molecular targeted therapy (BRAF inhibitor + MEK inhibitor) have revolutionized the management of adjuvant treatment for melanoma. As the results from these trials will mature in the next years, a change in the landscape of adjuvant treatment for melanoma is expected, resulting in new challenges in treatment decisions such as optimizing patients' selection through predictive and prognostic biomarkers, and management of treatment related adverse events, in particular immune related toxicities.
Collapse
Affiliation(s)
- S Napolitano
- Oncologia Medica, Dipartimento di Internistica Clinica e Sperimentale "F. Magrassi", Università degli Studi della Campania "Luigi Vanvitelli", Via S. Pansini 5, Napoli 80131, Italy
| | - G Brancaccio
- Dermatologia e Venerologia, Dipartimento di salute mentale e fisica e medicina riabilitativa, Università degli Studi della Campania "Luigi Vanvitelli", Via S. Pansini 5, Napoli 80131, Italy
| | - G Argenziano
- Dermatologia e Venerologia, Dipartimento di salute mentale e fisica e medicina riabilitativa, Università degli Studi della Campania "Luigi Vanvitelli", Via S. Pansini 5, Napoli 80131, Italy
| | - E Martinelli
- Oncologia Medica, Dipartimento di Internistica Clinica e Sperimentale "F. Magrassi", Università degli Studi della Campania "Luigi Vanvitelli", Via S. Pansini 5, Napoli 80131, Italy
| | - F Morgillo
- Oncologia Medica, Dipartimento di Internistica Clinica e Sperimentale "F. Magrassi", Università degli Studi della Campania "Luigi Vanvitelli", Via S. Pansini 5, Napoli 80131, Italy
| | - F Ciardiello
- Oncologia Medica, Dipartimento di Internistica Clinica e Sperimentale "F. Magrassi", Università degli Studi della Campania "Luigi Vanvitelli", Via S. Pansini 5, Napoli 80131, Italy
| | - T Troiani
- Oncologia Medica, Dipartimento di Internistica Clinica e Sperimentale "F. Magrassi", Università degli Studi della Campania "Luigi Vanvitelli", Via S. Pansini 5, Napoli 80131, Italy.
| |
Collapse
|
2624
|
Wan M, Ming M. Nivolumab versus ipilimumab in the treatment of advanced melanoma: a critical appraisal. Br J Dermatol 2018; 179:296-300. [DOI: 10.1111/bjd.16785] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- M.T. Wan
- Department of Dermatology; University of Pennsylvania Perelman School of Medicine; Philadelphia PA U.S.A
| | - M.E. Ming
- Department of Dermatology; University of Pennsylvania Perelman School of Medicine; Philadelphia PA U.S.A
| |
Collapse
|
2625
|
Santoni M, Massari F, Di Nunno V, Conti A, Cimadamore A, Scarpelli M, Montironi R, Cheng L, Battelli N, Lopez-Beltran A. Immunotherapy in renal cell carcinoma: latest evidence and clinical implications. Drugs Context 2018; 7:212528. [PMID: 29899754 PMCID: PMC5992965 DOI: 10.7573/dic.212528] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 04/11/2018] [Accepted: 04/16/2018] [Indexed: 12/12/2022] Open
Abstract
Advances in understanding the mechanisms of tumour-induced immunosuppression have led to the development of immune-checkpoint inhibitors in cancer patients, including those with renal cell carcinoma (RCC). The optimal combination between immunotherapy and targeted agents (as well as the possible favourable sequential therapy of these two classes of drugs) remains an open question at this moment. Several trials are currently underway to assess the combination of anti-programmed-death 1 (PD-1) or anti-PD-ligand(L)1 agents with other immunotherapies or with anti-vascular endothelial growth factor receptor (VEGFR) tyrosine kinase inhibitors (TKIs). In this editorial, we described the results of the most recent clinical trials on the use of immunotherapies in RCC and the emerging data on the research for reliable biomarkers of tumour response in this setting. In addition, we have focused on the role of the gut microbiome and tumour microenvironment in the development of future therapeutic strategies for RCC patients.
Collapse
Affiliation(s)
| | | | | | - Alessandro Conti
- Department of Urology, Bressanone/Brixen Hospital, Bressanone, Italy
| | - Alessia Cimadamore
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, Ancona, Italy
| | - Marina Scarpelli
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, Ancona, Italy
| | - Rodolfo Montironi
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, Ancona, Italy
| | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Antonio Lopez-Beltran
- Department of Pathology and Surgery, University of Cordoba, Faculty of Medicine, Cordoba, Spain
| |
Collapse
|
2626
|
Morrison AH, Byrne KT, Vonderheide RH. Immunotherapy and Prevention of Pancreatic Cancer. Trends Cancer 2018; 4:418-428. [PMID: 29860986 PMCID: PMC6028935 DOI: 10.1016/j.trecan.2018.04.001] [Citation(s) in RCA: 323] [Impact Index Per Article: 46.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 03/30/2018] [Accepted: 04/03/2018] [Indexed: 12/16/2022]
Abstract
Pancreatic cancer is the third-leading cause of cancer mortality in the USA, recently surpassing breast cancer. A key component of pancreatic cancer's lethality is its acquired immune privilege, which is driven by an immunosuppressive microenvironment, poor T cell infiltration, and a low mutational burden. Although immunotherapies such as checkpoint blockade or engineered T cells have yet to demonstrate efficacy, a growing body of evidence suggests that orthogonal combinations of these and other strategies could unlock immunotherapy in pancreatic cancer. In this Review article, we discuss promising immunotherapies currently under investigation in pancreatic cancer and provide a roadmap for the development of prevention vaccines for this and other cancers.
Collapse
Affiliation(s)
- Alexander H Morrison
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19014, USA
| | - Katelyn T Byrne
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19014, USA; Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA 19014, USA
| | - Robert H Vonderheide
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19014, USA; Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA 19014, USA; Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19014, USA.
| |
Collapse
|
2627
|
Lee A, Sun S, Sandler A, Hoang T. Recent progress in therapeutic antibodies for cancer immunotherapy. Curr Opin Chem Biol 2018; 44:56-65. [DOI: 10.1016/j.cbpa.2018.05.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 05/03/2018] [Indexed: 12/19/2022]
|
2628
|
Advanced Melanoma: Current Treatment Options, Biomarkers, and Future Perspectives. Am J Clin Dermatol 2018; 19:303-317. [PMID: 29164492 DOI: 10.1007/s40257-017-0325-6] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Malignant melanoma accounts for the highest number of deaths from skin cancer, and the prognosis of patients with stage IV disease has historically been poor. Novel insights into both mutations driving tumorigenesis and immune escape mechanisms of these tumors have led to effective treatment options that have revolutionized the treatment of this disease. Targeting the MAPK kinase pathway (with BRAF and MEK inhibitors), as well as targeting checkpoints, such as cytotoxic T-lymphocyte associated protein 4 (CTLA-4) or programmed death 1 (PD-1), have improved overall survival in patients with late-stage melanoma, and biomarker research for personalized therapy is ongoing for each of these treatment modalities. In this review, we will discuss current first-line treatment options, discuss biomarkers supporting treatment decisions, and give an outlook on (combination) therapies we expect to become relevant in the near future.
Collapse
|
2629
|
Suzman DL, Pelosof L, Rosenberg A, Avigan MI. Hepatotoxicity of immune checkpoint inhibitors: An evolving picture of risk associated with a vital class of immunotherapy agents. Liver Int 2018; 38:976-987. [PMID: 29603856 DOI: 10.1111/liv.13746] [Citation(s) in RCA: 157] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 03/21/2018] [Indexed: 12/13/2022]
Abstract
Immune checkpoint inhibitors (ICIs) block CTLA-4, PD-1 and PD-L1, or other molecules that control antitumour activities of lymphocytes. These products are associated with a broad array of immune-related toxicities affecting a variety of organs, including the liver. ICI-associated immune-mediated hepatitis (IMH) ranges in severity between mild and life-threatening and is marked by findings that bear both similarities as well as differences with idiopathic autoimmune hepatitis. Hepatotoxic events are often detected in clinical trials of ICIs that are powered for efficacy. Risk levels for ICI-induced liver injury may be impacted by the specific checkpoint molecule targeted for treatment, the ICI dose levels, and the presence of a pre-existing autoimmune diathesis, chronic infection or tumour cells which infiltrate the liver parenchyma. When patients develop liver injury during ICI treatment, a prompt assessment of the cause of injury, in conjunction with the application of measures to optimally manage the adverse event, should be made. Strategies to manage the risk of IMH include the performance of pretreatment liver tests with regular monitoring during and after ICI treatment and patient education. Using Common Terminology Criteria for Adverse Events developed at the National Cancer Institute to measure the severity level of liver injury, recommended actions may include continued ICI treatment with close patient monitoring, ICI treatment suspension or discontinuation and/or administration of corticosteroids or, when necessary, a non-steroidal immunosuppressive agent. The elucidation of reliable predictors of tumour-specific ICI treatment responses, as well as an increased susceptibility for clinically serious immune-related adverse events, would help optimize treatment decisions for individual patients.
Collapse
Affiliation(s)
- Daniel L Suzman
- Office of Hematology and Oncology Products, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Lorraine Pelosof
- Office of Hematology and Oncology Products, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Amy Rosenberg
- Office of Biotechnology Products, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Mark I Avigan
- Office of Surveillance and Epidemiology, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| |
Collapse
|
2630
|
Sullivan RJ, Atkins MB, Kirkwood JM, Agarwala SS, Clark JI, Ernstoff MS, Fecher L, Gajewski TF, Gastman B, Lawson DH, Lutzky J, McDermott DF, Margolin KA, Mehnert JM, Pavlick AC, Richards JM, Rubin KM, Sharfman W, Silverstein S, Slingluff CL, Sondak VK, Tarhini AA, Thompson JA, Urba WJ, White RL, Whitman ED, Hodi FS, Kaufman HL. An update on the Society for Immunotherapy of Cancer consensus statement on tumor immunotherapy for the treatment of cutaneous melanoma: version 2.0. J Immunother Cancer 2018; 6:44. [PMID: 29848375 PMCID: PMC5977556 DOI: 10.1186/s40425-018-0362-6] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 05/17/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Cancer immunotherapy has been firmly established as a standard of care for patients with advanced and metastatic melanoma. Therapeutic outcomes in clinical trials have resulted in the approval of 11 new drugs and/or combination regimens for patients with melanoma. However, prospective data to support evidence-based clinical decisions with respect to the optimal schedule and sequencing of immunotherapy and targeted agents, how best to manage emerging toxicities and when to stop treatment are not yet available. METHODS To address this knowledge gap, the Society for Immunotherapy of Cancer (SITC) Melanoma Task Force developed a process for consensus recommendations for physicians treating patients with melanoma integrating evidence-based data, where available, with best expert consensus opinion. The initial consensus statement was published in 2013, and version 2.0 of this report is an update based on a recent meeting of the Task Force and extensive subsequent discussions on new agents, contemporary peer-reviewed literature and emerging clinical data. The Academy of Medicine (formerly Institute of Medicine) clinical practice guidelines were used as a basis for consensus development with an updated literature search for important studies published between 1992 and 2017 and supplemented, as appropriate, by recommendations from Task Force participants. RESULTS The Task Force considered patients with stage II-IV melanoma and here provide consensus recommendations for how they would incorporate the many immunotherapy options into clinical pathways for patients with cutaneous melanoma. CONCLUSION These clinical guidleines provide physicians and healthcare providers with consensus recommendations for managing melanoma patients electing treatment with tumor immunotherapy.
Collapse
Affiliation(s)
- Ryan J. Sullivan
- Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114 USA
| | | | | | - Sanjiv S. Agarwala
- St. Luke’s Cancer Center and Temple University, Center Valley, PA 18034 USA
| | | | | | | | | | | | | | - Jose Lutzky
- Mt. Sinai Medical Center, Miami Beach, FL 33140 USA
| | | | | | | | - Anna C. Pavlick
- New York University Cancer Institute, New York, NY 10016 USA
| | | | - Krista M. Rubin
- Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114 USA
| | - William Sharfman
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21231 USA
| | | | | | - Vernon K. Sondak
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612 USA
| | | | | | - Walter J. Urba
- Earle A. Chiles Research Institute, Providence Cancer Center, Portland, OR 97213 USA
| | | | | | | | - Howard L. Kaufman
- Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114 USA
| |
Collapse
|
2631
|
Gershenwald JE, Scolyer RA. Melanoma Staging: American Joint Committee on Cancer (AJCC) 8th Edition and Beyond. Ann Surg Oncol 2018; 25:2105-2110. [PMID: 29850954 DOI: 10.1245/s10434-018-6513-7] [Citation(s) in RCA: 318] [Impact Index Per Article: 45.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Indexed: 12/23/2022]
Affiliation(s)
- Jeffrey E Gershenwald
- Departments of Surgical Oncology and Cancer Biology, Unit 1484, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. .,Melanoma and Skin Center, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Richard A Scolyer
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia.,Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia.,Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
2632
|
Forget MA, Haymaker C, Hess KR, Meng YJ, Creasy C, Karpinets T, Fulbright OJ, Roszik J, Woodman SE, Kim YU, Sakellariou-Thompson D, Bhatta A, Wahl A, Flores E, Thorsen ST, Tavera RJ, Ramachandran R, Gonzalez AM, Toth CL, Wardell S, Mansaray R, Patel V, Carpio DJ, Vaughn C, Farinas CM, Velasquez PG, Hwu WJ, Patel SP, Davies MA, Diab A, Glitza IC, Tawbi H, Wong MK, Cain S, Ross MI, Lee JE, Gershenwald JE, Lucci A, Royal R, Cormier JN, Wargo JA, Radvanyi LG, Torres-Cabala CA, Beroukhim R, Hwu P, Amaria RN, Bernatchez C. Prospective Analysis of Adoptive TIL Therapy in Patients with Metastatic Melanoma: Response, Impact of Anti-CTLA4, and Biomarkers to Predict Clinical Outcome. Clin Cancer Res 2018; 24:4416-4428. [PMID: 29848573 DOI: 10.1158/1078-0432.ccr-17-3649] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 04/11/2018] [Accepted: 05/23/2018] [Indexed: 11/16/2022]
Abstract
Purpose: Adoptive cell therapy (ACT) using tumor-infiltrating lymphocytes (TIL) has consistently demonstrated clinical efficacy in metastatic melanoma. Recent widespread use of checkpoint blockade has shifted the treatment landscape, raising questions regarding impact of these therapies on response to TIL and appropriate immunotherapy sequence.Patients and Methods: Seventy-four metastatic melanoma patients were treated with autologous TIL and evaluated for clinical response according to irRC, overall survival, and progression-free survival. Immunologic factors associated with response were also evaluated.Results: Best overall response for the entire cohort was 42%; 47% in 43 checkpoint-naïve patients, 38% when patients were exposed to anti-CTLA4 alone (21 patients) and 33% if also exposed to anti-PD1 (9 patients) prior to TIL ACT. Median overall survival was 17.3 months; 24.6 months in CTLA4-naïve patients and 8.6 months in patients with prior CTLA4 blockade. The latter patients were infused with fewer TIL and experienced a shorter duration of response. Infusion of higher numbers of TIL with CD8 predominance and expression of BTLA correlated with improved response in anti-CTLA4 naïve patients, but not in anti-CTLA4 refractory patients. Baseline serum levels of IL9 predicted response to TIL ACT, while TIL persistence, tumor recognition, and mutation burden did not correlate with outcome.Conclusions: This study demonstrates the deleterious effects of prior exposure to anti-CTLA4 on TIL ACT response and shows that baseline IL9 levels can potentially serve as a predictive tool to select the appropriate sequence of immunotherapies. Clin Cancer Res; 24(18); 4416-28. ©2018 AACR.
Collapse
Affiliation(s)
- Marie-Andrée Forget
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center (MDACC), Houston, Texas
| | - Cara Haymaker
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center (MDACC), Houston, Texas
| | - Kenneth R Hess
- Department of Biostatistics, The University of Texas MDACC, Houston, Texas
| | - Yuzhong Jeff Meng
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts.,Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Caitlin Creasy
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center (MDACC), Houston, Texas
| | - Tatiana Karpinets
- Department of Genomic Medicine, The University of Texas MDACC, Houston, Texas
| | - Orenthial J Fulbright
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center (MDACC), Houston, Texas
| | - Jason Roszik
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center (MDACC), Houston, Texas.,Department of Genomic Medicine, The University of Texas MDACC, Houston, Texas
| | - Scott E Woodman
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center (MDACC), Houston, Texas
| | - Young Uk Kim
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center (MDACC), Houston, Texas
| | | | - Ankit Bhatta
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center (MDACC), Houston, Texas
| | - Arely Wahl
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center (MDACC), Houston, Texas
| | - Esteban Flores
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center (MDACC), Houston, Texas
| | - Shawne T Thorsen
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center (MDACC), Houston, Texas
| | - René J Tavera
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center (MDACC), Houston, Texas
| | - Renjith Ramachandran
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center (MDACC), Houston, Texas
| | - Audrey M Gonzalez
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center (MDACC), Houston, Texas
| | - Christopher L Toth
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center (MDACC), Houston, Texas
| | - Seth Wardell
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center (MDACC), Houston, Texas
| | - Rahmatu Mansaray
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center (MDACC), Houston, Texas
| | - Vruti Patel
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center (MDACC), Houston, Texas
| | - Destiny Joy Carpio
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center (MDACC), Houston, Texas
| | - Carol Vaughn
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center (MDACC), Houston, Texas
| | - Chantell M Farinas
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center (MDACC), Houston, Texas
| | - Portia G Velasquez
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center (MDACC), Houston, Texas
| | - Wen-Jen Hwu
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center (MDACC), Houston, Texas
| | - Sapna P Patel
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center (MDACC), Houston, Texas
| | - Michael A Davies
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center (MDACC), Houston, Texas
| | - Adi Diab
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center (MDACC), Houston, Texas
| | - Isabella C Glitza
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center (MDACC), Houston, Texas
| | - Hussein Tawbi
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center (MDACC), Houston, Texas
| | - Michael K Wong
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center (MDACC), Houston, Texas
| | - Suzanne Cain
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center (MDACC), Houston, Texas
| | - Merrick I Ross
- Department of Surgical Oncology, The University of Texas MDACC, Houston, Texas
| | - Jeffrey E Lee
- Department of Surgical Oncology, The University of Texas MDACC, Houston, Texas
| | | | - Anthony Lucci
- Department of Surgical Oncology, The University of Texas MDACC, Houston, Texas
| | - Richard Royal
- Department of Surgical Oncology, The University of Texas MDACC, Houston, Texas
| | - Janice N Cormier
- Department of Surgical Oncology, The University of Texas MDACC, Houston, Texas
| | - Jennifer A Wargo
- Department of Genomic Medicine, The University of Texas MDACC, Houston, Texas.,Department of Surgical Oncology, The University of Texas MDACC, Houston, Texas
| | - Laszlo G Radvanyi
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center (MDACC), Houston, Texas
| | | | - Rameen Beroukhim
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts.,Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Patrick Hwu
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center (MDACC), Houston, Texas
| | - Rodabe N Amaria
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center (MDACC), Houston, Texas.
| | - Chantale Bernatchez
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center (MDACC), Houston, Texas.
| |
Collapse
|
2633
|
Chen H, Chen Y, Liu H, Que Y, Zhang X, Zheng F. Integrated Expression Profiles Analysis Reveals Correlations Between the IL-33/ST2 Axis and CD8 + T Cells, Regulatory T Cells, and Myeloid-Derived Suppressor Cells in Soft Tissue Sarcoma. Front Immunol 2018; 9:1179. [PMID: 29896199 PMCID: PMC5986931 DOI: 10.3389/fimmu.2018.01179] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 05/11/2018] [Indexed: 01/07/2023] Open
Abstract
Soft tissue sarcoma (STS) is a rare solid malignant cancer, and there are few effective treatment options for advanced disease. Cancer immunotherapy is a promising new strategy for STS treatment. IL-33 is a candidate cytokine for immunotherapy that can activate T lymphocytes and modulate antitumor immunity in some cancers. However, the expression and biological role of IL-33 in STS are poorly understood. In this study, we found that the expression of IL-33 and its receptor ST2 was decreased in STS using real-time PCR assays. By analyzing sarcoma data from The Cancer Genome Atlas, we found that higher transcriptional levels of IL-33 and ST2 were associated with a favorable outcome. There were positive correlations between the expression levels of ST2 and CD3E, CD4, CD8A, CD45RO, FOXP3, CD11B, CD33, and IFN-γ. Strong positive correlations between the expression of IFN-γ and CD3E and CD8A were also observed. Moreover, the expression levels of both IL-33 and ST2 were positively correlated with those of CD3E, CD8A, and chemokines that recruit CD8+ T cells, indicating that the IL-33/ST2 axis may play an important role in recruiting and promoting the immune response of type 1-polarized CD8+ T cells in STS. Meanwhile, we also found that the expression of IL-33 was negatively correlated with that of TGF-β1 and chemokines that recruit regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs), indicating that the IL-33/ST2 axis may also contribute to antagonizing Tregs, MDSCs, and TGF-β1-mediated immunosuppression in STS. The correlations between the IL-33/ST2 axis and CD8+ T cells and IFN-γ, as well as Tregs, MDSCs, and TGF-β1 were validated by additional analyses using three other independent GEO datasets of sarcoma. Our results implicate the possible role of the IL-33/ST2 axis in modulating antitumor immunity in STS. IL-33 may not only serve as a useful prognostic biomarker for STS but also as a potential therapeutic target for STS immunotherapy and worth further investigation.
Collapse
Affiliation(s)
- Huoying Chen
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guilin Medical University, Guilin, China.,Melanoma and Sarcoma Medical Oncology Unit, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.,Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yao Chen
- Melanoma and Sarcoma Medical Oncology Unit, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.,Department of Radiotherapy, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Hongbo Liu
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Yi Que
- Melanoma and Sarcoma Medical Oncology Unit, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xing Zhang
- Melanoma and Sarcoma Medical Oncology Unit, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Fang Zheng
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| |
Collapse
|
2634
|
Hogan SA, Levesque MP, Cheng PF. Melanoma Immunotherapy: Next-Generation Biomarkers. Front Oncol 2018; 8:178. [PMID: 29896449 PMCID: PMC5986946 DOI: 10.3389/fonc.2018.00178] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 05/08/2018] [Indexed: 01/05/2023] Open
Abstract
The recent emergence of cancer immunotherapies initiated a significant shift in the clinical management of metastatic melanoma. Prior to 2011, melanoma patients only had palliative treatment solutions which offered little to no survival benefit. In 2018, with immunotherapy, melanoma patients can now contemplate durable or even complete remission. Treatment with novel immune checkpoint inhibitors, anti-cytotoxic T-lymphocyte protein 4 and anti-programmed cell death protein 1, clearly result in superior median and long-term survivals compared to standard chemotherapy; however, more than half of the patients do not respond to immune checkpoint blockade. Currently, clinicians do not have any effective way to stratify melanoma patients for immunotherapies. Research is now focusing on identifying biomarkers which could predict a patient’s response prior treatment initiation (or very early during treatment course), in order to maximize therapeutic efficacy, avoid unnecessary costs, and undesirable heavy side effects for the patient. Given the rapid developments in this field and the translational potential for some of the biomarkers, we will summarize the current state of biomarker research for immunotherapy in melanoma, with an emphasis on omics technologies such as next-generation sequencing and mass cytometry (CyTOF).
Collapse
Affiliation(s)
- Sabrina A Hogan
- Department of Dermatology, UniversitätsSpital Zürich, Gloriastrasse, Zurich, Switzerland.,Faculty of Medicine, Universität Zürich, Zürich, Switzerland
| | - Mitchell P Levesque
- Department of Dermatology, UniversitätsSpital Zürich, Gloriastrasse, Zurich, Switzerland.,Faculty of Medicine, Universität Zürich, Zürich, Switzerland
| | - Phil F Cheng
- Department of Dermatology, UniversitätsSpital Zürich, Gloriastrasse, Zurich, Switzerland.,Faculty of Medicine, Universität Zürich, Zürich, Switzerland
| |
Collapse
|
2635
|
Hilmi M, Bartholin L, Neuzillet C. Immune therapies in pancreatic ductal adenocarcinoma: Where are we now? World J Gastroenterol 2018; 24:2137-2151. [PMID: 29853732 PMCID: PMC5974576 DOI: 10.3748/wjg.v24.i20.2137] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 05/05/2018] [Accepted: 05/18/2018] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers, mostly due to its resistance to treatment. Of these, checkpoint inhibitors (CPI) are inefficient when used as monotherapy, except in the case of a rare subset of tumors harboring microsatellite instability (< 2%). This inefficacy mainly resides in the low immunogenicity and non-inflamed phenotype of PDAC. The abundant stroma generates a hypoxic microenvironment and drives the recruitment of immunosuppressive cells through cancer-associated-fibroblast activation and transforming growth factor β secretion. Several strategies have recently been developed to overcome this immunosuppressive microenvironment. Combination therapies involving CPI aim at increasing tumor immunogenicity and promoting the recruitment and activation of effector T cells. Ongoing studies are therefore exploring the association of CPI with vaccines, oncolytic viruses, MEK inhibitors, cytokine inhibitors, and hypoxia- and stroma-targeting agents. Adoptive T-cell transfer is also under investigation. Moreover, translational studies on tumor tissue and blood, prior to and during treatment may lead to the identification of biomarkers with predictive value for both clinical outcome and response to immunotherapy.
Collapse
Affiliation(s)
- Marc Hilmi
- Service d’Oncologie Médicale, Hôpital Henri Mondor, Assistance Publique-Hôpitaux de Paris, Université Paris Est Créteil, Créteil 94010, France
| | - Laurent Bartholin
- Université de Lyon, Université Claude Bernard Lyon 1, Inserm U1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon 69008, France
| | - Cindy Neuzillet
- Service d’Oncologie Médicale, Hôpital Henri Mondor, Assistance Publique-Hôpitaux de Paris, Université Paris Est Créteil, Créteil 94010, France
| |
Collapse
|
2636
|
Effective Immunotherapy in Bone Marrow Metastatic Melanoma Presenting with Disseminated Intravascular Coagulopathy. Case Reports Immunol 2018; 2018:4520294. [PMID: 29796327 PMCID: PMC5896417 DOI: 10.1155/2018/4520294] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 12/27/2017] [Indexed: 12/26/2022] Open
Abstract
Malignant melanoma is responsible for the majority of skin cancer deaths and is increasing in prevalence. Bone marrow (BM) involvement by melanoma is rare in the absence of widespread visceral disease. Here, we report the case of a 30-year-old female who presented to the hospital with back pain, low-grade fever, and easy bruising. She was found to be bicytopenic and in disseminated intravascular coagulopathy (DIC). Surprisingly, BM biopsy showed extensive involvement by metastatic malignant melanoma in the absence of visceral or brain metastasis. The unique presentation of this case and the challenge of management of a potentially treatable cancer in a critically ill patient are discussed, alongside a review of published cases of metastatic melanoma in the BM and an exploration of currently available treatment options. The excellent response of our patient to combined immune checkpoint inhibitors has yet to be paralleled in the available literature.
Collapse
|
2637
|
LeBlanc TW, Temel JS, Helft PR. "How Much Time Do I Have?": Communicating Prognosis in the Era of Exceptional Responders. Am Soc Clin Oncol Educ Book 2018; 38:787-794. [PMID: 30231384 DOI: 10.1200/edbk_201211] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Prognostication is the science by which clinicians estimate a patient's expected outcome. A robust literature shows that many patients with advanced cancer have inaccurate perceptions of their prognosis, thus raising questions about whether patients are truly making informed decisions. Clinicians' ability to communicate prognostic information is further complicated today by the availability of novel, efficacious immunotherapies and genome-guided treatments. Currently, clinicians lack tools to predict which patients with advanced disease will achieve an exceptional response to these new therapies. This increased prognostic uncertainty on the part of clinicians further complicates prognostic communication with patients. Evidence also suggests that many oncologists avoid or rarely engage in prognosis-related communication and/or lack skills in this area. Although communication skills training interventions can have a positive impact on complex communication skills for some clinicians, there is no one-size-fits-all approach to improving patient-clinician communication about prognosis. Yet improving patient understanding of prognosis is critical, because patient understanding of prognosis is linked with end-of-life care outcomes. Solutions to this problem will likely require a combination of interventions beyond communication skills training programs, including enhanced use of other cancer clinicians, such as oncology nurses and social workers, increased use of palliative care specialists, and organizational support to facilitate advance care planning.
Collapse
Affiliation(s)
- Thomas W LeBlanc
- From the Cancer Patient Experience Research Program, Duke Cancer Institute, Division of Hematologic Malignancies and Cellular Therapy, Duke University School of Medicine, Durham, NC; Massachusetts General Hospital, Boston, MA; Department of Medicine, Division of Hematology/Oncology, Indiana University Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN
| | - Jennifer S Temel
- From the Cancer Patient Experience Research Program, Duke Cancer Institute, Division of Hematologic Malignancies and Cellular Therapy, Duke University School of Medicine, Durham, NC; Massachusetts General Hospital, Boston, MA; Department of Medicine, Division of Hematology/Oncology, Indiana University Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN
| | - Paul R Helft
- From the Cancer Patient Experience Research Program, Duke Cancer Institute, Division of Hematologic Malignancies and Cellular Therapy, Duke University School of Medicine, Durham, NC; Massachusetts General Hospital, Boston, MA; Department of Medicine, Division of Hematology/Oncology, Indiana University Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN
| |
Collapse
|
2638
|
Läubli H, Balmelli C, Kaufmann L, Stanczak M, Syedbasha M, Vogt D, Hertig A, Müller B, Gautschi O, Stenner F, Zippelius A, Egli A, Rothschild SI. Influenza vaccination of cancer patients during PD-1 blockade induces serological protection but may raise the risk for immune-related adverse events. J Immunother Cancer 2018; 6:40. [PMID: 29789020 PMCID: PMC5964701 DOI: 10.1186/s40425-018-0353-7] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 05/09/2018] [Indexed: 12/12/2022] Open
Abstract
Background Immune checkpoint inhibiting antibodies were introduced into routine clinical practice for cancer patients. Checkpoint blockade has led to durable remissions in some patients, but may also induce immune-related adverse events (irAEs). Lung cancer patients show an increased risk for complications, when infected with influenza viruses. Therefore, vaccination is recommended. However, the efficacy and safety of influenza vaccination during checkpoint blockade and its influence on irAEs is unclear. Similarly, the influence of vaccinations on T cell-mediated immune reactions in patients during PD-1 blockade remains poorly defined. Methods We vaccinated 23 lung cancer patients and 11 age-matched healthy controls using a trivalent inactivated influenza vaccine to investigate vaccine-induced immunity and safety during checkpoint blockade. Results We did not observe significant differences between patients and healthy controls in vaccine-induced antibody titers against all three viral antigens. Influenza vaccination resulted in protective titers in more than 60% of patients/participants. In cancer patients, the post-vaccine frequency of irAEs was 52.2% with a median time to occurrence of 3.2 months after vaccination. Six of 23 patients (26.1%) showed severe grade 3/4 irAEs. This frequency of irAEs might be higher than the rate previously published in the literature and the rate observed in a non-study population at our institution (all grades 25.5%, grade 3/4 9.8%). Conclusions Although this is a non-randomized trial with a limited number of patients, the increased rate of immunological toxicity is concerning. This finding should be studied in a larger patient population.
Collapse
Affiliation(s)
- Heinz Läubli
- Department of Internal Medicine, Division of Medical Oncology, University Hospital Basel, Basel, Switzerland.,Cancer Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Catharina Balmelli
- Department of Internal Medicine, Division of Medical Oncology, University Hospital Basel, Basel, Switzerland
| | - Lukas Kaufmann
- Applied Microbiology Research, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Michal Stanczak
- Cancer Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Mohammedyaseen Syedbasha
- Applied Microbiology Research, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Dominik Vogt
- Applied Microbiology Research, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Astrid Hertig
- Department of Internal Medicine, Division of Medical Oncology, University Hospital Basel, Basel, Switzerland
| | - Beat Müller
- Oncology, Cantonal Hospital Lucerne, Lucerne, Switzerland
| | | | - Frank Stenner
- Department of Internal Medicine, Division of Medical Oncology, University Hospital Basel, Basel, Switzerland.,Cancer Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Alfred Zippelius
- Department of Internal Medicine, Division of Medical Oncology, University Hospital Basel, Basel, Switzerland.,Cancer Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Adrian Egli
- Applied Microbiology Research, Department of Biomedicine, University of Basel, Basel, Switzerland.,Clinical Microbiology, University Hospital Basel, Basel, Switzerland
| | - Sacha I Rothschild
- Department of Internal Medicine, Division of Medical Oncology, University Hospital Basel, Basel, Switzerland. .,Cancer Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland.
| |
Collapse
|
2639
|
Baas P, Disselhorst M. Immuno-oncology in malignant pleural mesothelioma. THE LANCET. RESPIRATORY MEDICINE 2018; 6:408-410. [PMID: 29773327 DOI: 10.1016/s2213-2600(18)30176-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Accepted: 04/19/2018] [Indexed: 12/23/2022]
Affiliation(s)
- Paul Baas
- Department of Thoracic Oncology, The Netherlands Cancer Institute, Amsterdam 1066CX, Netherlands.
| | - Maria Disselhorst
- Department of Thoracic Oncology, The Netherlands Cancer Institute, Amsterdam 1066CX, Netherlands
| |
Collapse
|
2640
|
Lin SY, Huang SK, Huynh KT, Salomon MP, Chang SC, Marzese DM, Lanman RB, Talasaz A, Hoon DS. Multiplex Gene Profiling of Cell-Free DNA in Patients With Metastatic Melanoma for Monitoring Disease. JCO Precis Oncol 2018; 2:PO.17.00225. [PMID: 32913981 PMCID: PMC7446321 DOI: 10.1200/po.17.00225] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
PURPOSE Hotspot blood cell-free DNA (cfDNA) biomarker assays have limited utility in profiling tumor heterogeneity and burden and in capturing regional metastasis with low disease burden in patients with melanoma. We investigated the utility of a sensitive 54-cancer gene digital next-generation sequencing approach targeting blood cfDNA single nucleotide variants (SNVs) and copy number amplification for monitoring disease in patients with melanoma with regional or distant organ metastasis (DOM). PATIENTS AND METHODS A total of 142 blood samples were evaluated by digital next-generation sequencing across two patient cohorts. Cohort 1 contained 44 patients with stage II, III, or IV disease with matched tumor DNA at the time of surgery or DOM. Cohort 2 consisted of 12 overlapping patients who were longitudinally monitored after complete lymph node dissection to DOM. RESULTS In cohort 1, cfDNA SNVs were detected in 75% of patients. Tumor-cfDNA somatic SNV concordance was 85% at a variant allele fraction of ≥ 0.5%. An SNV load (number of unique SNVs detected) of greater than two SNVs and an SNV burden (total cumulative SNV VAF) of > 0.5% were significantly associated with worse overall survival (P < .05) in stage IV patients. In cohort 2, 98 longitudinal blood samples along with matched regional and distant metastases from 12 stage III patients were analyzed before complete lymph node dissection and throughout disease progression. cfDNA SNV levels correlated with tumor burden (P = .019), enabled earlier detection of recurrence compared with radiologic imaging (P < .01), captured tumor heterogeneity, and identified increasing SNVs levels before recurrence. CONCLUSION This study demonstrates significant utility for cfDNA profiling in patients with melanoma with regional and/or distant metastasis for earlier detection of recurrence and progression and in capturing tumor evolution and heterogeneity, thus impacting how patients with melanoma are monitored.
Collapse
Affiliation(s)
- Selena Y. Lin
- Selena Y. Lin, Sharon K. Huang, Kelly T. Huynh, Matthew P. Salomon, Diego M. Marzese, and Dave S.B. Hoon, John Wayne Cancer Institute at Providence Saint John’s Health Center, Santa Monica; Richard B. Lanman and AmirAli Talasaz, Guardant Health, Redwood City, CA; and Shu-Ching Chang, Medical Data Research Center at Providence Saint Joseph’s Health, Portland, OR
| | - Sharon K. Huang
- Selena Y. Lin, Sharon K. Huang, Kelly T. Huynh, Matthew P. Salomon, Diego M. Marzese, and Dave S.B. Hoon, John Wayne Cancer Institute at Providence Saint John’s Health Center, Santa Monica; Richard B. Lanman and AmirAli Talasaz, Guardant Health, Redwood City, CA; and Shu-Ching Chang, Medical Data Research Center at Providence Saint Joseph’s Health, Portland, OR
| | - Kelly T. Huynh
- Selena Y. Lin, Sharon K. Huang, Kelly T. Huynh, Matthew P. Salomon, Diego M. Marzese, and Dave S.B. Hoon, John Wayne Cancer Institute at Providence Saint John’s Health Center, Santa Monica; Richard B. Lanman and AmirAli Talasaz, Guardant Health, Redwood City, CA; and Shu-Ching Chang, Medical Data Research Center at Providence Saint Joseph’s Health, Portland, OR
| | - Matthew P. Salomon
- Selena Y. Lin, Sharon K. Huang, Kelly T. Huynh, Matthew P. Salomon, Diego M. Marzese, and Dave S.B. Hoon, John Wayne Cancer Institute at Providence Saint John’s Health Center, Santa Monica; Richard B. Lanman and AmirAli Talasaz, Guardant Health, Redwood City, CA; and Shu-Ching Chang, Medical Data Research Center at Providence Saint Joseph’s Health, Portland, OR
| | - Shu-Ching Chang
- Selena Y. Lin, Sharon K. Huang, Kelly T. Huynh, Matthew P. Salomon, Diego M. Marzese, and Dave S.B. Hoon, John Wayne Cancer Institute at Providence Saint John’s Health Center, Santa Monica; Richard B. Lanman and AmirAli Talasaz, Guardant Health, Redwood City, CA; and Shu-Ching Chang, Medical Data Research Center at Providence Saint Joseph’s Health, Portland, OR
| | - Diego M. Marzese
- Selena Y. Lin, Sharon K. Huang, Kelly T. Huynh, Matthew P. Salomon, Diego M. Marzese, and Dave S.B. Hoon, John Wayne Cancer Institute at Providence Saint John’s Health Center, Santa Monica; Richard B. Lanman and AmirAli Talasaz, Guardant Health, Redwood City, CA; and Shu-Ching Chang, Medical Data Research Center at Providence Saint Joseph’s Health, Portland, OR
| | - Richard B. Lanman
- Selena Y. Lin, Sharon K. Huang, Kelly T. Huynh, Matthew P. Salomon, Diego M. Marzese, and Dave S.B. Hoon, John Wayne Cancer Institute at Providence Saint John’s Health Center, Santa Monica; Richard B. Lanman and AmirAli Talasaz, Guardant Health, Redwood City, CA; and Shu-Ching Chang, Medical Data Research Center at Providence Saint Joseph’s Health, Portland, OR
| | - AmirAli Talasaz
- Selena Y. Lin, Sharon K. Huang, Kelly T. Huynh, Matthew P. Salomon, Diego M. Marzese, and Dave S.B. Hoon, John Wayne Cancer Institute at Providence Saint John’s Health Center, Santa Monica; Richard B. Lanman and AmirAli Talasaz, Guardant Health, Redwood City, CA; and Shu-Ching Chang, Medical Data Research Center at Providence Saint Joseph’s Health, Portland, OR
| | - Dave S.B. Hoon
- Selena Y. Lin, Sharon K. Huang, Kelly T. Huynh, Matthew P. Salomon, Diego M. Marzese, and Dave S.B. Hoon, John Wayne Cancer Institute at Providence Saint John’s Health Center, Santa Monica; Richard B. Lanman and AmirAli Talasaz, Guardant Health, Redwood City, CA; and Shu-Ching Chang, Medical Data Research Center at Providence Saint Joseph’s Health, Portland, OR
| |
Collapse
|
2641
|
Chen CB, Wu MY, Ng CY, Lu CW, Wu J, Kao PH, Yang CK, Peng MT, Huang CY, Chang WC, Hui RCY, Yang CH, Yang SF, Chung WH, Su SC. Severe cutaneous adverse reactions induced by targeted anticancer therapies and immunotherapies. Cancer Manag Res 2018; 10:1259-1273. [PMID: 29844705 PMCID: PMC5962313 DOI: 10.2147/cmar.s163391] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
With the increasing use of targeted anticancer drugs and immunotherapies, there have been a substantial number of reports concerning life-threatening severe cutaneous adverse reactions (SCARs), including Stevens–Johnson syndrome (SJS), toxic epidermal necrolysis (TEN), drug rash with eosinophilia and systemic symptoms, drug-induced hypersensitivity syndrome, and acute generalized exanthematous pustulosis. Although the potential risks and characteristics for targeted anticancer agent- and immunotherapy-induced SCAR were not well understood, these serious adverse reactions usually result in morbidity and sequela. As a treatment guideline for this devastating condition is still unavailable, prompt withdrawal of causative drugs is believed to be a priority of patient management. In this review, we outline distinct types of SCARs caused by targeted anticancer therapies and immunotherapies. Also, we discuss the clinical course, latency, concomitant medication, tolerability of rechallenge or alternatives, tumor response, and mortality associated with these devastating conditions. Imatinib, vemurafenib, and rituximab were the top three offending medications that most commonly caused SJS/TEN, while EGFR inhibitors were the group of drugs that most frequently induced SJS/TEN. For drug rash with eosinophilia and systemic symptoms/drug-induced hypersensitivity syndrome and acute generalized exanthematous pustulosis, imatinib was also the most common offending drug. Additionally, we delineated 10 SCAR cases related to innovative immunotherapies, including PD1 and CTLA4 inhibitors. There was a wide range of latency periods: 5.5–91 days (median). Only eight of 16 reported patients with SCAR showed clinical responses. Targeted anticancer drugs and immunotherapies can lead to lethal SCAR (14 deceased patients were identified as suffering from SJS/TEN). The mortality rate of TEN was high: up to 52.4%. The information compiled herein will serve as a solid foundation to formulate ideas for early recognition of SCAR and to discontinue offending drugs for better management.
Collapse
Affiliation(s)
- Chun-Bing Chen
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Linkou, Taiwan.,Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Taipei, Taiwan.,Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Keelung, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Immune-Oncology Center of Excellence, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Ming-Ying Wu
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Linkou, Taiwan.,Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Taipei, Taiwan.,Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Keelung, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chau Yee Ng
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Linkou, Taiwan.,Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Taipei, Taiwan.,Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Keelung, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chun-Wei Lu
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Linkou, Taiwan.,Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Taipei, Taiwan.,Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Keelung, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Immune-Oncology Center of Excellence, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Jennifer Wu
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Linkou, Taiwan.,Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Taipei, Taiwan.,Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Keelung, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Immune-Oncology Center of Excellence, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Pei-Han Kao
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Linkou, Taiwan.,Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Taipei, Taiwan.,Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Keelung, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Immune-Oncology Center of Excellence, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Chan-Keng Yang
- College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Immune-Oncology Center of Excellence, Chang Gung Memorial Hospital, Linkou, Taiwan.,Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Meng-Ting Peng
- College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Immune-Oncology Center of Excellence, Chang Gung Memorial Hospital, Linkou, Taiwan.,Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Chen-Yang Huang
- College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Immune-Oncology Center of Excellence, Chang Gung Memorial Hospital, Linkou, Taiwan.,Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Wen-Cheng Chang
- College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Immune-Oncology Center of Excellence, Chang Gung Memorial Hospital, Linkou, Taiwan.,Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Rosaline Chung-Yee Hui
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Linkou, Taiwan.,Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Taipei, Taiwan.,Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Keelung, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chih-Hsun Yang
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Linkou, Taiwan.,Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Taipei, Taiwan.,Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Keelung, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Wen-Hung Chung
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Linkou, Taiwan.,Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Taipei, Taiwan.,Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Keelung, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Immune-Oncology Center of Excellence, Chang Gung Memorial Hospital, Linkou, Taiwan.,Whole-Genome Research Core Laboratory of Human Diseases, Chang Gung Memorial Hospital, Keelung, Taiwan.,Department of Dermatology, Xiamen Chang Gung Hospital, Xiamen, China
| | - Shih-Chi Su
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Linkou, Taiwan.,Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Taipei, Taiwan.,Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Keelung, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Whole-Genome Research Core Laboratory of Human Diseases, Chang Gung Memorial Hospital, Keelung, Taiwan
| |
Collapse
|
2642
|
|
2643
|
Chae YK, Arya A, Iams W, Cruz MR, Chandra S, Choi J, Giles F. Current landscape and future of dual anti-CTLA4 and PD-1/PD-L1 blockade immunotherapy in cancer; lessons learned from clinical trials with melanoma and non-small cell lung cancer (NSCLC). J Immunother Cancer 2018; 6:39. [PMID: 29769148 PMCID: PMC5956851 DOI: 10.1186/s40425-018-0349-3] [Citation(s) in RCA: 308] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Accepted: 05/02/2018] [Indexed: 12/14/2022] Open
Abstract
Immunotherapy is among the most rapidly evolving treatment strategies in oncology. The therapeutic potential of immune-checkpoint inhibitors is exemplified by the recent hail of Food and Drug Administration (FDA) approvals for their use in various malignancies. Continued efforts to enhance outcomes with immunotherapy agents have led to the formulation of advanced treatment strategies. Recent evidence from pre-clinical studies evaluating immune-checkpoint inhibitors in various cancer cell-lines has suggested that combinatorial approaches may have superior survival outcomes compared to single-agent immunotherapy regimens. Preliminary trials assessing combination therapy with anti-PD-1/PD-L1 plus anti-CTLA-4 immune-checkpoint inhibitors have documented considerable advantages in survival indices over single-agent immunotherapy. The therapeutic potential of combinatorial approaches is highlighted by the recent FDA approval of nivolumab plus ipilimumab for patients with advanced melanoma. Presently, dual-immune checkpoint inhibition with anti-programmed death receptor-1/programmed cell death receptor- ligand-1 (anti-PD-1/PD-L1) plus anti-cytotoxic T lymphocyte associated antigen-4 (anti-CTLA-4) monoclonal antibodies (MoAbs) is being evaluated for a wide range of tumor histologies. Furthermore, several ongoing clinical trials are investigating combination checkpoint inhibition in association with traditional treatment modalities such as chemotherapy, surgery, and radiation. In this review, we summarize the current landscape of combination therapy with anti-PD-1/PD-L1 plus anti-CTLA-4 MoAbs for patients with melanoma and non-small cell lung cancer (NSCLC). We present a synopsis of the prospects for expanding the indications of dual immune-checkpoint inhibition therapy to a more diverse set of tumor histologies.
Collapse
Affiliation(s)
- Young Kwang Chae
- Developmental Therapeutics Program of the Division of Hematology Oncology, Early Phase Clinical Trials Unit, 645 N. Michigan Avenue, Suite 1006, Chicago, IL, 60611, USA. .,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, 645 N. Michigan Avenue, Suite 1006, Chicago, IL, 60611, USA. .,Northwestern University Feinberg School of Medicine, 645 N. Michigan Avenue, Suite 1006, Chicago, IL, 60611, USA.
| | - Ayush Arya
- Developmental Therapeutics Program of the Division of Hematology Oncology, Early Phase Clinical Trials Unit, 645 N. Michigan Avenue, Suite 1006, Chicago, IL 60611 USA
| | - Wade Iams
- 0000 0001 2299 3507grid.16753.36Northwestern University Feinberg School of Medicine, 645 N. Michigan Avenue, Suite 1006, Chicago, IL 60611 USA
| | - Marcelo R. Cruz
- Developmental Therapeutics Program of the Division of Hematology Oncology, Early Phase Clinical Trials Unit, 645 N. Michigan Avenue, Suite 1006, Chicago, IL 60611 USA
| | - Sunandana Chandra
- Developmental Therapeutics Program of the Division of Hematology Oncology, Early Phase Clinical Trials Unit, 645 N. Michigan Avenue, Suite 1006, Chicago, IL 60611 USA ,0000 0001 2299 3507grid.16753.36Robert H. Lurie Comprehensive Cancer Center of Northwestern University, 645 N. Michigan Avenue, Suite 1006, Chicago, IL 60611 USA ,0000 0001 2299 3507grid.16753.36Northwestern University Feinberg School of Medicine, 645 N. Michigan Avenue, Suite 1006, Chicago, IL 60611 USA
| | - Jaehyuk Choi
- 0000 0001 2299 3507grid.16753.36Robert H. Lurie Comprehensive Cancer Center of Northwestern University, 645 N. Michigan Avenue, Suite 1006, Chicago, IL 60611 USA ,0000 0001 2299 3507grid.16753.36Northwestern University Feinberg School of Medicine, 645 N. Michigan Avenue, Suite 1006, Chicago, IL 60611 USA
| | - Francis Giles
- Developmental Therapeutics Program of the Division of Hematology Oncology, Early Phase Clinical Trials Unit, 645 N. Michigan Avenue, Suite 1006, Chicago, IL 60611 USA ,0000 0001 2299 3507grid.16753.36Robert H. Lurie Comprehensive Cancer Center of Northwestern University, 645 N. Michigan Avenue, Suite 1006, Chicago, IL 60611 USA ,0000 0001 2299 3507grid.16753.36Northwestern University Feinberg School of Medicine, 645 N. Michigan Avenue, Suite 1006, Chicago, IL 60611 USA
| |
Collapse
|
2644
|
Wang PF, Cai HQ, Zhang CB, Li YM, Liu X, Wan JH, Jiang T, Li SW, Yan CX. Molecular and clinical characterization of PTPN2 expression from RNA-seq data of 996 brain gliomas. J Neuroinflammation 2018; 15:145. [PMID: 29764444 PMCID: PMC5953404 DOI: 10.1186/s12974-018-1187-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 05/03/2018] [Indexed: 12/14/2022] Open
Abstract
Background Immune checkpoint inhibitors have been shown to promote antitumor immunity and achieve durable tumor remissions. However, certain tumors are refractory to current immunotherapy. These negative results encouraged us to uncover other therapeutic targets and strategies. PTPN2 (protein tyrosine phosphatase, non-receptor type 2) has been newly identified as an immunotherapy target. Loss of PTPN2 sensitizes the tumor to immunotherapy via IFNγ signaling. Methods Here, we investigated the relationship between PTPN2 mRNA levels and clinical characteristics in gliomas. RNA-seq data of a cohort of 325 patients with glioma were available from the Chinese Glioma Genome Atlas and 671 from The Cancer Genome Atlas. R language, GraphPad Prism 5, and SPSS 22.0 were used to analyze data and draw figures. Results PTPN2 transcript levels increased significantly with higher grades of glioma and in isocitrate dehydrogenase (IDH) wild-type and mesenchymal subtype gliomas. A comprehensive biological analysis was conducted, which indicated a crucial role of PTPN2 in the immune and inflammation responses in gliomas. Specifically, PTPN2 was positively associated with HCK, LCK, MHC II, and STAT1 but negatively related to IgG and interferon. Moreover, canonical correlation analysis showed a positive correlation of PTPN2 with infiltrating immune cells, such as macrophages, neutrophils, and CD8+ T cells. Clinically, higher levels of PTPN2 were associated with a worse overall survival both in patients with gliomas and glioblastomas. Conclusion PTPN2 expression level was increased in glioblastomas and associated with gliomas of the IDH wild-type and mesenchymal subtype. There was a close correlation between PTPN2 and the immune response and inflammatory activity in gliomas. Our results show that PTPN2 is a promising immunotherapy target and may provide additional treatment strategies. Electronic supplementary material The online version of this article (10.1186/s12974-018-1187-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Peng-Fei Wang
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Building 1, Ward 6, Xiang Shan Yi Ke Song Road 50, Haidian, Beijing, China
| | - Hong-Qing Cai
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Building 1, Ward 6, Xiang Shan Yi Ke Song Road 50, Haidian, Beijing, China.,Department of Neurosurgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chuan-Bao Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Beijing Neurosurgical Institute, Beijing, China.,Chinese Glioma Genome Atlas Network (CGGA), Beijing, China
| | - Yan-Michael Li
- Department of Neurosurgery and Oncology, University of Rochester Medical Center, Rochester, NY, USA
| | - Xiang Liu
- Department of Imaging Sciences, University of Rochester Medical Center, Rochester, NY, USA
| | - Jing-Hai Wan
- Department of Neurosurgery, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tao Jiang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Beijing Neurosurgical Institute, Beijing, China.,Chinese Glioma Genome Atlas Network (CGGA), Beijing, China
| | - Shou-Wei Li
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Building 1, Ward 6, Xiang Shan Yi Ke Song Road 50, Haidian, Beijing, China.
| | - Chang-Xiang Yan
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Building 1, Ward 6, Xiang Shan Yi Ke Song Road 50, Haidian, Beijing, China.
| |
Collapse
|
2645
|
Hellmann MD, Nathanson T, Rizvi H, Creelan BC, Sanchez-Vega F, Ahuja A, Ni A, Novik JB, Mangarin LMB, Abu-Akeel M, Liu C, Sauter JL, Rekhtman N, Chang E, Callahan MK, Chaft JE, Voss MH, Tenet M, Li XM, Covello K, Renninger A, Vitazka P, Geese WJ, Borghaei H, Rudin CM, Antonia SJ, Swanton C, Hammerbacher J, Merghoub T, McGranahan N, Snyder A, Wolchok JD. Genomic Features of Response to Combination Immunotherapy in Patients with Advanced Non-Small-Cell Lung Cancer. Cancer Cell 2018; 33:843-852.e4. [PMID: 29657128 PMCID: PMC5953836 DOI: 10.1016/j.ccell.2018.03.018] [Citation(s) in RCA: 799] [Impact Index Per Article: 114.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Revised: 02/09/2018] [Accepted: 03/16/2018] [Indexed: 12/29/2022]
Abstract
Combination immune checkpoint blockade has demonstrated promising benefit in lung cancer, but predictors of response to combination therapy are unknown. Using whole-exome sequencing to examine non-small-cell lung cancer (NSCLC) treated with PD-1 plus CTLA-4 blockade, we found that high tumor mutation burden (TMB) predicted improved objective response, durable benefit, and progression-free survival. TMB was independent of PD-L1 expression and the strongest feature associated with efficacy in multivariable analysis. The low response rate in TMB low NSCLCs demonstrates that combination immunotherapy does not overcome the negative predictive impact of low TMB. This study demonstrates the association between TMB and benefit to combination immunotherapy in NSCLC. TMB should be incorporated in future trials examining PD-(L)1 with CTLA-4 blockade in NSCLC.
Collapse
Affiliation(s)
- Matthew D Hellmann
- Department of Medicine, Memorial Sloan Kettering Cancer Center, 885 2(nd) Avenue, New York, NY 10017, USA; Weill Cornell School of Medicine, New York, NY, USA; Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Tavi Nathanson
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hira Rizvi
- Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Benjamin C Creelan
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Francisco Sanchez-Vega
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Marie-Josèe and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Arun Ahuja
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ai Ni
- Department of Biostatistics and Epidemiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jacki B Novik
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Levi M B Mangarin
- Ludwig Collaborative Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mohsen Abu-Akeel
- Ludwig Collaborative Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Cailian Liu
- Ludwig Collaborative Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jennifer L Sauter
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Natasha Rekhtman
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Eliza Chang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Margaret K Callahan
- Department of Medicine, Memorial Sloan Kettering Cancer Center, 885 2(nd) Avenue, New York, NY 10017, USA; Weill Cornell School of Medicine, New York, NY, USA; Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jamie E Chaft
- Department of Medicine, Memorial Sloan Kettering Cancer Center, 885 2(nd) Avenue, New York, NY 10017, USA; Weill Cornell School of Medicine, New York, NY, USA; Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Martin H Voss
- Department of Medicine, Memorial Sloan Kettering Cancer Center, 885 2(nd) Avenue, New York, NY 10017, USA; Weill Cornell School of Medicine, New York, NY, USA
| | - Megan Tenet
- Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Xue-Mei Li
- Bristol Myers Squibb, Princeton, NJ, USA
| | | | | | | | | | | | - Charles M Rudin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, 885 2(nd) Avenue, New York, NY 10017, USA; Weill Cornell School of Medicine, New York, NY, USA; Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Scott J Antonia
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Charles Swanton
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK; Translational Cancer Therapeutics Laboratory, Francis Crick Institute, London, UK
| | - Jeff Hammerbacher
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Taha Merghoub
- Department of Medicine, Memorial Sloan Kettering Cancer Center, 885 2(nd) Avenue, New York, NY 10017, USA; Weill Cornell School of Medicine, New York, NY, USA; Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Ludwig Collaborative Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nicholas McGranahan
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Alexandra Snyder
- Department of Medicine, Memorial Sloan Kettering Cancer Center, 885 2(nd) Avenue, New York, NY 10017, USA
| | - Jedd D Wolchok
- Department of Medicine, Memorial Sloan Kettering Cancer Center, 885 2(nd) Avenue, New York, NY 10017, USA; Weill Cornell School of Medicine, New York, NY, USA; Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Ludwig Collaborative Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
2646
|
Kantarjian HM, Prat F, Steensma DP, Kurzrock R, Stewart DJ, Sekeres MA, Leveque J. Cancer research in the United States: A critical review of current status and proposal for alternative models. Cancer 2018; 124:2881-2889. [DOI: 10.1002/cncr.31522] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 02/27/2018] [Accepted: 03/28/2018] [Indexed: 12/18/2022]
Affiliation(s)
- Hagop M. Kantarjian
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Ferran Prat
- Research Administration and Industry Ventures; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - David P. Steensma
- Department of Medical Oncology; Dana-Farber Cancer Institute, Harvard Medical School; Boston Massachusetts
| | - Razelle Kurzrock
- Division of Hematology/Oncology, Department of Medicine; University of California at San Diego; San Diego California
| | - David J. Stewart
- Department of Medicine; University of Ottawa; Ottawa Ontario Canada
| | | | | |
Collapse
|
2647
|
Konieczkowski DJ, Johannessen CM, Garraway LA. A Convergence-Based Framework for Cancer Drug Resistance. Cancer Cell 2018; 33:801-815. [PMID: 29763622 PMCID: PMC5957297 DOI: 10.1016/j.ccell.2018.03.025] [Citation(s) in RCA: 168] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 02/02/2018] [Accepted: 03/26/2018] [Indexed: 02/07/2023]
Abstract
Despite advances in cancer biology and therapeutics, drug resistance remains problematic. Resistance is often multifactorial, heterogeneous, and prone to undersampling. Nonetheless, many individual mechanisms of targeted therapy resistance may coalesce into a smaller number of convergences, including pathway reactivation (downstream re-engagement of original effectors), pathway bypass (recruitment of a parallel pathway converging on the same downstream output), and pathway indifference (development of a cellular state independent of the initial therapeutic target). Similar convergences may also underpin immunotherapy resistance. Such parsimonious, convergence-based frameworks may help explain resistance across tumor types and therapeutic categories and may also suggest strategies to overcome it.
Collapse
|
2648
|
Balch CM. Revolutionary Advances in Immunotherapy for Melanoma Are Coming into the Surgical Arena: Are We Ready? Ann Surg Oncol 2018; 25:1803-1806. [PMID: 29752603 DOI: 10.1245/s10434-018-6516-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Indexed: 01/07/2023]
Affiliation(s)
- Charles M Balch
- University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
2649
|
Wang Y, Abu-Sbeih H, Mao E, Ali N, Ali FS, Qiao W, Lum P, Raju G, Shuttlesworth G, Stroehlein J, Diab A. Immune-checkpoint inhibitor-induced diarrhea and colitis in patients with advanced malignancies: retrospective review at MD Anderson. J Immunother Cancer 2018; 6:37. [PMID: 29747688 PMCID: PMC5946546 DOI: 10.1186/s40425-018-0346-6] [Citation(s) in RCA: 182] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 04/26/2018] [Indexed: 12/11/2022] Open
Abstract
Background Immune checkpoint inhibitors (ICPIs) are gaining increasing popularity as an efficacious treatment for advanced malignancies. ICPI treatment can be complicated by diarrhea and colitis. Systemic steroids are the first line treatment. Infliximab is reserved for severe refractory cases. We aimed to assess the impact of ICPI-induced diarrhea and colitis and their immunosuppressive treatment on patients’ outcomes. Methods This retrospective analysis was conducted in 327 cancer patients who received ICPIs between 2011 and 2017. Patients with ICPI-induced toxicities in other organs were excluded. We collected data about patient demographics, clinical variables, and overall survival. We used descriptive analysis to compare different groups based on the occurrence and the treatment of diarrhea and colitis. Kaplan-Meier and log-rank test were used to estimate and compare overall survival durations between groups. Results Diarrhea was recorded in 117 (36%) patients; 79 (24%) of them required immunosuppressive treatment of either systemic corticosteroid without infliximab (n = 44) or with infliximab (n = 35). Caucasian ethnicity, melanoma, stage 3 cancer, and ipilimumab were predictors of colitis that requires immunosuppression. Patients who required immunosuppressants had better overall survival than those who did not require treatment for colitis or diarrhea (P < 0.001). Immunosuppression for diarrhea or colitis did not affect the overall survival significantly (P = 0.232), nor did the choice of treatment (corticosteroids with vs. without infliximab; P = 0.768). Diarrhea was an independent predictor of a favorable overall survival (P < 0.001), irrespective of treatment need (P = 0.003). We confirmed the same results in a subgroup analysis for patients with stage IV malignancies only. Patients who received long duration of steroid treatment (> 30 days) had numerically higher infection rate than those who received steroid for shorter duration (40.4 vs. 25.8%, P = 0.160). Likewise, long duration of steroid without infliximab was associated with increased risk of infection compared to short duration of steroid with infliximab (42.9% vs. 14.3%, P = 0.089). Conclusions Patients with ICPI-induced diarrhea or colitis have improved survival outcomes. Diarrhea is an independent predictor of an improved survival regardless of treatment requirement. Immunosuppressive treatment for diarrhea did not significantly affect overall survival, however, infection rates were numerically higher among patients who received steroids for a long duration. Therefore, early non-steroid immunosuppressive therapy may ensure a more favorable overall outcome.
Collapse
Affiliation(s)
- Yinghong Wang
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.
| | - Hamzah Abu-Sbeih
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Emily Mao
- Department of Internal Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Noman Ali
- Department of Internal Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Faisal Shaukat Ali
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Wei Qiao
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Phillip Lum
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Gottumukkala Raju
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Gladis Shuttlesworth
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - John Stroehlein
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Adi Diab
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
2650
|
Morrison C, Pabla S, Conroy JM, Nesline MK, Glenn ST, Dressman D, Papanicolau-Sengos A, Burgher B, Andreas J, Giamo V, Qin M, Wang Y, Lenzo FL, Omilian A, Bshara W, Zibelman M, Ghatalia P, Dragnev K, Shirai K, Madden KG, Tafe LJ, Shah N, Kasuganti D, de la Cruz-Merino L, Araujo I, Saenger Y, Bogardus M, Villalona-Calero M, Diaz Z, Day R, Eisenberg M, Anderson SM, Puzanov I, Galluzzi L, Gardner M, Ernstoff MS. Predicting response to checkpoint inhibitors in melanoma beyond PD-L1 and mutational burden. J Immunother Cancer 2018; 6:32. [PMID: 29743104 PMCID: PMC5944039 DOI: 10.1186/s40425-018-0344-8] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 04/20/2018] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have changed the clinical management of melanoma. However, not all patients respond, and current biomarkers including PD-L1 and mutational burden show incomplete predictive performance. The clinical validity and utility of complex biomarkers have not been studied in melanoma. METHODS Cutaneous metastatic melanoma patients at eight institutions were evaluated for PD-L1 expression, CD8+ T-cell infiltration pattern, mutational burden, and 394 immune transcript expression. PD-L1 IHC and mutational burden were assessed for association with overall survival (OS) in 94 patients treated prior to ICI approval by the FDA (historical-controls), and in 137 patients treated with ICIs. Unsupervised analysis revealed distinct immune-clusters with separate response rates. This comprehensive immune profiling data were then integrated to generate a continuous Response Score (RS) based upon response criteria (RECIST v.1.1). RS was developed using a single institution training cohort (n = 48) and subsequently tested in a separate eight institution validation cohort (n = 29) to mimic a real-world clinical scenario. RESULTS PD-L1 positivity ≥1% correlated with response and OS in ICI-treated patients, but demonstrated limited predictive performance. High mutational burden was associated with response in ICI-treated patients, but not with OS. Comprehensive immune profiling using RS demonstrated higher sensitivity (72.2%) compared to PD-L1 IHC (34.25%) and tumor mutational burden (32.5%), but with similar specificity. CONCLUSIONS In this study, the response score derived from comprehensive immune profiling in a limited melanoma cohort showed improved predictive performance as compared to PD-L1 IHC and tumor mutational burden.
Collapse
Affiliation(s)
- Carl Morrison
- Center for Personalized Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA.
- Department of Pathology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA.
- Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA.
- OmniSeq Inc., Buffalo, NY, 14203, USA.
| | | | - Jeffrey M Conroy
- Center for Personalized Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
- OmniSeq Inc., Buffalo, NY, 14203, USA
| | | | - Sean T Glenn
- Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
- OmniSeq Inc., Buffalo, NY, 14203, USA
| | | | | | | | | | | | | | | | | | - Angela Omilian
- Department of Pathology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Wiam Bshara
- Department of Pathology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Matthew Zibelman
- Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA
| | - Pooja Ghatalia
- Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA
| | - Konstantin Dragnev
- Department of Hematology and Oncology, Dartmouth Hitchcock, Lebanon, NH, 03756, USA
| | - Keisuke Shirai
- Department of Hematology and Oncology, Dartmouth Hitchcock, Lebanon, NH, 03756, USA
| | - Katherine G Madden
- Department of Hematology and Oncology, Dartmouth Hitchcock, Lebanon, NH, 03756, USA
| | - Laura J Tafe
- Department of Hematology and Oncology, Dartmouth Hitchcock, Lebanon, NH, 03756, USA
- Department of Pathology, Dartmouth Hitchcock, Lebanon, NH, 03756, USA
| | - Neel Shah
- Department of Pathology, Community Hospital, Munster, IN, 46321, USA
| | - Deepa Kasuganti
- Department of Pathology, Community Hospital, Munster, IN, 46321, USA
| | - Luis de la Cruz-Merino
- Department of Clinical Oncology Development, Hospital Universitario Virgen Macarena, 41009, Sevilla, Spain
| | - Isabel Araujo
- Department of Clinical Oncology Development, Hospital Universitario Virgen Macarena, 41009, Sevilla, Spain
| | - Yvonne Saenger
- Department of Medicine, Columbia University, New York, NY, 10032, USA
| | - Margaret Bogardus
- Department of Medicine, Columbia University, New York, NY, 10032, USA
| | | | - Zuanel Diaz
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, 33176, USA
| | - Roger Day
- Department of Biomedical Informatics and Biostatistics, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Marcia Eisenberg
- Laboratory Corporation of America Holdings, Burlington, NC, 27215, USA
| | - Steven M Anderson
- Laboratory Corporation of America Holdings, Burlington, NC, 27215, USA
| | - Igor Puzanov
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, 10065, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, 10065, USA
- Université Paris Descartes/Paris V, 75006, Paris, France
| | | | - Marc S Ernstoff
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| |
Collapse
|