3201
|
Onesti CE, Frères P, Jerusalem G. Atypical patterns of response to immune checkpoint inhibitors: interpreting pseudoprogression and hyperprogression in decision making for patients' treatment. J Thorac Dis 2019; 11:35-38. [PMID: 30863564 DOI: 10.21037/jtd.2018.12.47] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Concetta Elisa Onesti
- Laboratory of Human Genetics, GIGA Research Institute, University of Liège, Liège, Belgium.,Department of Medical Oncology, University Hospital (CHU Liège), Liège, Belgium.,Laboratory of Medical Oncology, GIGA Research Institute, University of Liège, Liège, Belgium
| | - Pierre Frères
- Department of Medical Oncology, University Hospital (CHU Liège), Liège, Belgium
| | - Guy Jerusalem
- Department of Medical Oncology, University Hospital (CHU Liège), Liège, Belgium.,Laboratory of Medical Oncology, GIGA Research Institute, University of Liège, Liège, Belgium
| |
Collapse
|
3202
|
Schmidt EV. Developing combination strategies using PD-1 checkpoint inhibitors to treat cancer. Semin Immunopathol 2019; 41:21-30. [PMID: 30374524 PMCID: PMC6323091 DOI: 10.1007/s00281-018-0714-9] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 09/18/2018] [Indexed: 02/08/2023]
Abstract
More than 3000 clinical trials are evaluating the clinical activity of the PD-1 checkpoint inhibitors as monotherapies and in combinations with other cancer therapies [1]. The PD-1 checkpoint inhibitors are remarkable for their clinical activities in shrinking tumors across a wide range of tumor types, in causing durable responses, and in their tolerability. These attributes position them as favorable agents in clinical combinations. Historically, approaches to cancer therapy combinations focused on agents with orthogonal activities to avoid shared resistance mechanisms and shared toxicities. Although CTLA-4/PD-1 combinations have progressed based on possible immune interactions, additional approaches have used more orthogonal treatments such as standard of care chemotherapies and anti-angiogenesis inhibitors. Using the concept of independent activity pioneered by Bliss [2], examples of these approaches were compared. Both standard of care chemotherapy and anti-angiogenesis combinations show promising clinical activity above that predicted by the independent contributions of the agents tested on their own. In contrast, the combinations of CTLA4/PD-1 checkpoint inhibitors in renal cancer and melanoma show no more activity than that predicted by the independent contributions of the monotherapies. This update on approaches to the development of clinical combination therapies highlights the potential importance of combining PD-1 checkpoint inhibitors with a broad range of clinically active partners.
Collapse
|
3203
|
Flippot R, Derosa L, Albiges L. Les métastases cérébrales de cancer du rein, un défi clinique. Bull Cancer 2019; 105 Suppl 3:S261-S267. [PMID: 30595155 DOI: 10.1016/s0007-4551(18)30381-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
BRAIN METASTASES IN RENAL CELL CARCINOMA, AN UNMET NEED Brain metastases from renal cell carcinomas are associated with dismal prognosis and might be present in up to 10 % of metastatic patients. Biologically, the blood brain barrier might be disrupted in brain metastases and thus do not exclusively account for treatment resistance. Brain metastases often acquire additional molecular alterations that might provide aggressive features. They are also associated with high lymphocytic infiltration and expression of immune checkpoints PD-1/PD-L1. In clinical routine, scores based on metastatic volume and patients' performance status might help better predict survival. The cornerstone of brain metastases treatment is stereotactic radiation therapy if patients are eligible, while systemic treatments such as antiangiogenics and immune checkpoint inhibitors only provide limited disease control. Early identification of patients with brain metastases from renal cell carcinomas and promotion of dedicated clinical trials will be important to try and improve current clinical management.
Collapse
Affiliation(s)
- Ronan Flippot
- Département de médecine oncologique, Gustave-Roussy, Villejuif, France
| | - Lisa Derosa
- Département de médecine oncologique, Gustave-Roussy, Villejuif, France
| | - Laurence Albiges
- Département de médecine oncologique, Gustave-Roussy, Villejuif, France.
| |
Collapse
|
3204
|
Elias R, Sharma A, Singla N, Brugarolas J. Next Generation Sequencing in Renal Cell Carcinoma: Towards Precision Medicine. KIDNEY CANCER JOURNAL : OFFICIAL JOURNAL OF THE KIDNEY CANCER ASSOCIATION 2019; 17:94-104. [PMID: 32206160 PMCID: PMC7089604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Affiliation(s)
- Roy Elias
- Department of Internal Medicine, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas TX, 75390
- Department of Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas TX, 75390
| | - Akanksha Sharma
- Department of Internal Medicine, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas TX, 75390
| | - Nirmish Singla
- Department of Urology, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas TX, 75390
| | - James Brugarolas
- Department of Internal Medicine, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas TX, 75390
- Department of Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas TX, 75390
| |
Collapse
|
3205
|
Chan TA, Yarchoan M, Jaffee E, Swanton C, Quezada SA, Stenzinger A, Peters S. Development of tumor mutation burden as an immunotherapy biomarker: utility for the oncology clinic. Ann Oncol 2019; 30:44-56. [PMID: 30395155 PMCID: PMC6336005 DOI: 10.1093/annonc/mdy495] [Citation(s) in RCA: 1849] [Impact Index Per Article: 308.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background Treatment with immune checkpoint blockade (ICB) with agents such as anti-programmed cell death protein 1 (PD-1), anti-programmed death-ligand 1 (PD-L1), and/or anti-cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) can result in impressive response rates and durable disease remission but only in a subset of patients with cancer. Expression of PD-L1 has demonstrated utility in selecting patients for response to ICB and has proven to be an important biomarker for patient selection. Tumor mutation burden (TMB) is emerging as a potential biomarker. However, refinement of interpretation and contextualization is required. Materials and methods In this review, we outline the evolution of TMB as a biomarker in oncology, delineate how TMB can be applied in the clinic, discuss current limitations as a diagnostic test, and highlight mechanistic insights unveiled by the study of TMB. We review available data to date studying TMB as a biomarker for response to ICB by tumor type, focusing on studies proposing a threshold for TMB as a predictive biomarker for ICB activity. Results High TMB consistently selects for benefit with ICB therapy. In lung, bladder and head and neck cancers, the current predictive TMB thresholds proposed approximate 200 non-synonymous somatic mutations by whole exome sequencing (WES). PD-L1 expression influences response to ICB in high TMB tumors with single agent PD-(L)1 antibodies; however, response may not be dependent on PD-L1 expression in the setting of anti-CTLA4 or anti-PD-1/CTLA-4 combination therapy. Disease-specific TMB thresholds for effective prediction of response in various other malignancies are not well established. Conclusions TMB, in concert with PD-L1 expression, has been demonstrated to be a useful biomarker for ICB selection across some cancer types; however, further prospective validation studies are required. TMB determination by selected targeted panels has been correlated with WES. Calibration and harmonization will be required for optimal utility and alignment across all platforms currently used internationally. Key challenges will need to be addressed before broader use in different tumor types.
Collapse
Affiliation(s)
- T A Chan
- Human Oncology and Pathogenesis Program; The Immunogenomics and Precision Oncology Program, Memorial Sloan Kettering Cancer Center, New York.
| | - M Yarchoan
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, USA
| | - E Jaffee
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, USA
| | - C Swanton
- Translational Cancer Therapeutics Laboratory, Francis Crick Institute, London
| | - S A Quezada
- Cancer Immunology Unit, Research Department of Haematology, University College London Cancer Institute, London, UK
| | - A Stenzinger
- Institute of Pathology, University Hospital Heidelberg, Heidelberg; Germany and German Cancer Consortium (DKTK), Heidelberg Partner Site, Heidelberg, Germany
| | - S Peters
- Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland.
| |
Collapse
|
3206
|
Berraondo P, Sanmamed MF, Ochoa MC, Etxeberria I, Aznar MA, Pérez-Gracia JL, Rodríguez-Ruiz ME, Ponz-Sarvise M, Castañón E, Melero I. Cytokines in clinical cancer immunotherapy. Br J Cancer 2019; 120:6-15. [PMID: 30413827 PMCID: PMC6325155 DOI: 10.1038/s41416-018-0328-y] [Citation(s) in RCA: 783] [Impact Index Per Article: 130.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 10/04/2018] [Accepted: 10/08/2018] [Indexed: 02/08/2023] Open
Abstract
Cytokines are soluble proteins that mediate cell-to-cell communication. Based on the discovery of the potent anti-tumour activities of several pro-inflammatory cytokines in animal models, clinical research led to the approval of recombinant interferon-alpha and interleukin-2 for the treatment of several malignancies, even if efficacy was only modest. These early milestones in immunotherapy have been followed by the recent addition to clinical practice of antibodies that inhibit immune checkpoints, as well as chimeric antigen receptor T cells. A renewed interest in the anti-tumour properties of cytokines has led to an exponential increase in the number of clinical trials that explore the safety and efficacy of cytokine-based drugs, not only as single agents, but also in combination with other immunomodulatory drugs. These second-generation drugs under clinical development include known molecules with novel mechanisms of action, new targets, and fusion proteins that increase half-life and target cytokine activity to the tumour microenvironment or to the desired effector immune cells. In addition, the detrimental activity of immunosuppressive cytokines can be blocked by antagonistic antibodies, small molecules, cytokine traps or siRNAs. In this review, we provide an overview of the novel trends in the cytokine immunotherapy field that are yielding therapeutic agents for clinical trials.
Collapse
Affiliation(s)
- Pedro Berraondo
- Immunology and Immunotherapy Program, Center for Applied Medical Research, CIMA, University of Navarra, Pamplona, Spain.
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Pamplona, Spain.
| | - Miguel F Sanmamed
- Immunology and Immunotherapy Program, Center for Applied Medical Research, CIMA, University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Pamplona, Spain
- Department of Oncology and immunology, Clínica Universidad de Navarra, Pamplona, Spain
| | - María C Ochoa
- Immunology and Immunotherapy Program, Center for Applied Medical Research, CIMA, University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Pamplona, Spain
| | - Iñaki Etxeberria
- Immunology and Immunotherapy Program, Center for Applied Medical Research, CIMA, University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Pamplona, Spain
| | - Maria A Aznar
- Immunology and Immunotherapy Program, Center for Applied Medical Research, CIMA, University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Pamplona, Spain
| | - José Luis Pérez-Gracia
- Immunology and Immunotherapy Program, Center for Applied Medical Research, CIMA, University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Pamplona, Spain
- Department of Oncology and immunology, Clínica Universidad de Navarra, Pamplona, Spain
| | - María E Rodríguez-Ruiz
- Immunology and Immunotherapy Program, Center for Applied Medical Research, CIMA, University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Pamplona, Spain
- Department of Oncology and immunology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Mariano Ponz-Sarvise
- Immunology and Immunotherapy Program, Center for Applied Medical Research, CIMA, University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Pamplona, Spain
- Department of Oncology and immunology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Eduardo Castañón
- Immunology and Immunotherapy Program, Center for Applied Medical Research, CIMA, University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Pamplona, Spain
- Department of Oncology and immunology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Ignacio Melero
- Immunology and Immunotherapy Program, Center for Applied Medical Research, CIMA, University of Navarra, Pamplona, Spain.
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Pamplona, Spain.
- Department of Oncology and immunology, Clínica Universidad de Navarra, Pamplona, Spain.
| |
Collapse
|
3207
|
Borchiellini D. Cancer du rein métastatique : quels critères de choix en 2e ligne ? Bull Cancer 2019; 105 Suppl 3:S242-S254. [PMID: 30595153 DOI: 10.1016/s0007-4551(18)30379-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
METASTATIC RENAL CELL CARCINOMA HOW TO CHOOSE THE APPROPRIATE SECOND-LINE TREATMENT?: The treatment of advanced or metastatic renal cell cancer (RCC) has dramatically improved in the past ten years. In the second-line setting, for patients who progressed on prior antiangiogenic therapy (mainly the VEGFR tyrosine kinase inhibitors (TKI) sunitinib or pazopanib), axitinib and everolimus have been recommended. Since 2015, other drugs have proven their efficacy and are currently considered the standard of care: cabozantinib (TKI that targets VEGFR, MET and AXL) and nivolumab (first anti-PD-1 check point inhibitor). Lenvatinib has also demonstrated promising results in association with everolimus, but this combination is not available in France. The optimal treatment choice for a given patient is challenging for the clinician when facing multiple options. In this article, we review the efficacy, safety and quality of life results of the main pivotal clinical studies involving advanced or metastatic RCC in the second-line setting, to help clinicians in selecting the most appropriate treatment. Beyond that, it is important to define all the sequencing strategy for patients to successively receive all the drugs that have demonstrated an increase in overall survival.
Collapse
Affiliation(s)
- Delphine Borchiellini
- Département d'oncologie médicale, centre Antoine-Lacassagne, université Côte d'Azur, Nice, France.
| |
Collapse
|
3208
|
Boilève A, Carlo MI, Barthélémy P, Oudard S, Borchiellini D, Voss MH, George S, Chevreau C, Landman-Parker J, Tabone MD, Chism DD, Amin A, Bilen MA, Bosse D, Coulomb-L'hermine A, Su X, Choueiri TK, Tannir NM, Malouf GG. Immune checkpoint inhibitors in MITF family translocation renal cell carcinomas and genetic correlates of exceptional responders. J Immunother Cancer 2018; 6:159. [PMID: 30591082 PMCID: PMC6307255 DOI: 10.1186/s40425-018-0482-z] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 12/13/2018] [Indexed: 01/05/2023] Open
Abstract
Background Microphthalmia Transcription Factor (MITF)family translocation renal cell carcinoma (tRCC) is a rare RCC subtype harboring TFE3/TFEB translocations. The prognosis in the metastatic (m) setting is poor. Programmed death ligand-1 expression was reported in 90% of cases, prompting us to analyze the benefit of immune checkpoint inhibitors (ICI) in this population. Patients and methods This multicenter retrospective study identified patients with MITF family mtRCC who had received an ICI in any of 12 referral centers in France or the USA. Response rate according to RECIST criteria, progression-free survival (PFS), and overall survival (OS) were analyzed. Genomic alterations associated with response were determined for 8 patients. Results Overall, 24 patients with metastatic disease who received an ICI as second or later line of treatment were identified. Nineteen (82.6%) of these patients had received a VEGFR inhibitor as first-line treatment, with a median PFS of 3 months (range, 1–22 months). The median PFS for patients during first ICI treatment was 2.5 months (range, 1–40 months); 4 patients experienced partial response (16,7%) and 3 (12,5%) had stable disease. Of the patients whose genomic alterations were analyzed, two patients with mutations in bromodomain-containing genes (PBRM1 and BRD8) had a clinical benefit. Resistant clones in a patient with exceptional response to ipilimumab showed loss of BRD8 mutations and increased mutational load driven by parallel evolution affecting 17 genes (median mutations per gene, 3), which were enriched mainly for O-glycan processing (29.4%, FDR = 9.7 × 10− 6). Conclusions MITF family tRCC is an aggressive disease with similar responses to ICIs as clear-cell RCC. Mutations in bromodomain-containing genes might be associated with clinical benefit. The unexpected observation about parallel evolution of genes involved in O-glycosylation as a mechanism of resistance to ICI warrants exploration.
Collapse
Affiliation(s)
- A Boilève
- Department of Medical Oncology, Hôpital Universitaire Pitié-Salpétrière, Paris, France
| | - M I Carlo
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - P Barthélémy
- Service d'Hématologie et d'Oncologie, Centre Hospitalier Universitaire de Strasbourg, Strasbourg, France
| | - S Oudard
- Oncology Department, European Georges Pompidou Hospital, René Descartes University, Paris, France.,Association pour la Recherche sur les Thérapeutiques Innovantes en Cancérologie, Paris, France.,U790 PARCC, European Georges Pompidou Hospital, René Descartes University, Paris, France
| | | | - M H Voss
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - S George
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - C Chevreau
- IUCT-Oncopole, Institut Claudius-Regaud, Toulouse, France
| | - J Landman-Parker
- Service d'Hématologie et d'Oncologie Pédiatrique, Hopital Armand-Trousseau, Paris, France
| | - M-D Tabone
- Service d'Hématologie et d'Oncologie Pédiatrique, Hopital Armand-Trousseau, Paris, France
| | - D D Chism
- Division of Hematology and Oncology, Department of Medicine, Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - A Amin
- Carolinas Healthcare System, Levine Cancer Institute, Charlotte, NC, USA
| | - M A Bilen
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - D Bosse
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Xiaoping Su
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - T K Choueiri
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Nizar M Tannir
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Gabriel G Malouf
- Department of Medical Oncology, Hôpital Universitaire Pitié-Salpétrière, Paris, France. .,Service d'Hématologie et d'Oncologie, Centre Hospitalier Universitaire de Strasbourg, Strasbourg, France. .,Department of Functional Genomics and Cancer, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France. .,Department of Hematology and Oncology, Centre Hospitalier Universitaire de Strasbourg, 1, Place de l'Hôpital, 67000, Strasbourg, France.
| |
Collapse
|
3209
|
Grünwald V, Hadaschik B. Re: Preliminary Results for Avelumab Plus Axitinib as First-Line Therapy in Patients with Advanced Clear-cell Renal-cell Carcinoma (JAVELIN Renal 100): An Open-label, Dose-finding and Dose-expansion, Phase 1b Trial. Eur Urol 2018; 75:697-698. [PMID: 30580900 DOI: 10.1016/j.eururo.2018.12.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 12/12/2018] [Indexed: 11/25/2022]
Affiliation(s)
- Viktor Grünwald
- Interdisciplinary Genitourinary Oncology, West-German Cancer Center, University Hospital Essen, Essen, Germany.
| | - Boris Hadaschik
- Department of Urology, University Hospital Essen, Essen, Germany
| |
Collapse
|
3210
|
Stitzlein L, Rao PSS, Dudley R. Emerging oral VEGF inhibitors for the treatment of renal cell carcinoma. Expert Opin Investig Drugs 2018; 28:121-130. [DOI: 10.1080/13543784.2019.1559296] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Lea Stitzlein
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Findlay, Findlay, OH, USA
| | - PSS Rao
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Findlay, Findlay, OH, USA
| | - Richard Dudley
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Findlay, Findlay, OH, USA
| |
Collapse
|
3211
|
Ansari J, Alhelali S, Albinmousa Z, Farrag A, Ali AM, Abdelgelil M, Alhamad A, Alzahrani G, Ansari A, Glaholm J. Rare Case of Intracardiac Renal Cell Carcinoma Metastasis with Response to Nivolumab: Case Report and Literature Review. Case Rep Oncol 2018; 11:861-870. [PMID: 30687063 PMCID: PMC6341359 DOI: 10.1159/000495459] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 11/13/2018] [Indexed: 12/28/2022] Open
Abstract
Intracardiac metastases in the absence of inferior vena cava involvement is a rare occurrence in patients with metastatic renal cell carcinoma (mRCC). There is limited evidence regarding the efficacy and safety of standard treatment modalities for mRCC patients with intracardiac metastases. Presence of intracardiac metastases is known to indicate poor prognosis and may potentially increase risk of treatment-related complications. Recent advances in RCC management have integrated nivolumab, a programmed death-1 (PD-1) receptor inhibitor, as a preferred treatment option in the second-line setting after failure of prior anti-angiogenic therapy; or in combination with ipilimumab, an anti-Cytotoxic T-lymphocyte antigen-4 antibody as first-line therapy for intermediate to poor risk patients with mRCC. The efficacy and toxicity of nivolumab in patients with mRCC and intracardiac metastases has never been reported previously. We herein present the first reported case of mRCC with intracardiac metastasis and a resultant excellent response to nivolumab treatment and discuss the imaging techniques and treatment options for this rare presentation.
Collapse
Affiliation(s)
- Jawaher Ansari
- Department of Oncology, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | | | | | - Ashraf Farrag
- Clinical Oncology Department, Assiut University, Asyut, Egypt
| | - Arwa M Ali
- Medical Oncology Department, South Egypt Cancer Institute, Asyut, Egypt
| | - Mai Abdelgelil
- Clinical Oncology Department, Assiut University, Asyut, Egypt
| | - Abdulaziz Alhamad
- Department of Oncology, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | | | - Asif Ansari
- Permian Basin Kidney Center, Odessa, Texas, USA
| | - John Glaholm
- Department of Oncology, Royal Marsden Hospital, London, United Kingdom
| |
Collapse
|
3212
|
Thrombotic Thrombocytopenic Purpura due to Checkpoint Inhibitors. Case Rep Hematol 2018; 2018:2464619. [PMID: 30671268 PMCID: PMC6317083 DOI: 10.1155/2018/2464619] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Revised: 11/23/2018] [Accepted: 11/28/2018] [Indexed: 12/28/2022] Open
Abstract
Ipilimumab is a monoclonal antibody that enhances the efficacy of the immune system by targeting a cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), which is a protein receptor that downregulates the immune system. Nivolumab is also a humanized monoclonal antibody that targets another protein receptor that prevents activated T cells from attacking the cancer; this receptor is called programmed cell death 1 (PD-1). The FDA approved ipilimumab combined with nivolumab as a frontline therapy for patients with metastatic melanoma or renal cell carcinoma and as a second-line therapy for patients with microsatellite instability-high (MSI-H) metastatic colon cancer. Immune-related adverse events such as autoimmune colitis, pneumonitis, hepatitis, nephritis, hypophysitis, and thyroiditis may occur during or weeks to months after therapy. We report a case of thrombotic thrombocytopenic purpura (TTP) in a patient with metastatic renal cell carcinoma following one cycle of ipilimumab and nivolumab. Only one case report of ipilimumab-induced TTP exists in the medical literature. With the wide use of immunotherapy to treat cancers, physicians need to be aware of this rare immune-related adverse event.
Collapse
|
3213
|
Park W, Lopes G. Perspectives: Neutrophil-to-lymphocyte Ratio as a Potential Biomarker in Immune Checkpoint Inhibitor for Non-Small-Cell Lung Cancer. Clin Lung Cancer 2018; 20:143-147. [PMID: 30683629 DOI: 10.1016/j.cllc.2018.12.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 11/25/2018] [Accepted: 12/08/2018] [Indexed: 12/18/2022]
Abstract
There is a rising need for optimal biomarkers to better tailor treatments for patients with cancer in the era of immunotherapy. In addition to programmed death-ligand 1 (PD-L1) and tumor mutation burden (TMB), neutrophil-to-lymphocyte (NLR) is regaining interest as a biomarker in immunotherapy for its availability, accessibility, and reproducibility. High NLR, according to different thresholds, is consistently reported to correlate with poor prognosis in different treatments in several cancers. Yet, most data come from retrospective analysis, and proof of mechanism and principle evaluations are limited. Prospective studies or adequately sized retrospective analyses of prospectively collected data are required to best assess its role in clinical practice. Moreover, effective myeloid or neutrophil modulators in tumor microenvironment can potentially contribute as a new therapeutic strategy. This perspective will summarize our current knowledge and will discuss where we stand now and propose future directions.
Collapse
Affiliation(s)
- Wungki Park
- Gastrointestinal Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY; Weill Cornell Medical College, New York, NY; Divisions of Hematology and Medical Oncology, Departments of Medicine, Miller School of Medicine, University of Miami, Sylvester Comprehensive Cancer Center, Miami, FL.
| | - Gilberto Lopes
- Divisions of Hematology and Medical Oncology, Departments of Medicine, Miller School of Medicine, University of Miami, Sylvester Comprehensive Cancer Center, Miami, FL
| |
Collapse
|
3214
|
Grassadonia A, Sperduti I, Vici P, Iezzi L, Brocco D, Gamucci T, Pizzuti L, Maugeri-Saccà M, Marchetti P, Cognetti G, De Tursi M, Natoli C, Barba M, Tinari N. Effect of Gender on the Outcome of Patients Receiving Immune Checkpoint Inhibitors for Advanced Cancer: A Systematic Review and Meta-Analysis of Phase III Randomized Clinical Trials. J Clin Med 2018; 7:jcm7120542. [PMID: 30545122 PMCID: PMC6306894 DOI: 10.3390/jcm7120542] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 11/30/2018] [Accepted: 12/08/2018] [Indexed: 12/26/2022] Open
Abstract
Evidence has recently emerged on the influence of gender on the immune system. In this systematic review and meta-analysis of phase III randomized clinical trials (RCTs), we explored the impact of gender on survival in patients with advanced cancer treated with immune checkpoint inhibitors (ICIs). We performed a comprehensive search of the literature updated to April 2018, including the Cochrane Central Register of Controlled Trials, PubMed, and EMBASE. We extracted data on study characteristics and risk of bias in duplicate. Of 423 unique citations, 21 RCTs were included, inherently to 12,635 patients. Both males and females showed reduced risk of death associated with ICIs use (HR 0.73, p < 0.001 and HR 0.77, p < 0.001, respectively). Subgroup analyses by specific ICI showed similar OS in both genders for anti-PD-1/PDL-1. Anti-CTLA-4 use was associated with longer OS in men only (HR 0.77, p < 0.012), with the exception of melanoma (in women, HR 0.80, p = 0.006). PFS was longer in men than in women (HR 0.67, p < 0.001 and HR 0.77, p = 0.100, respectively). Conclusively, ICIs use was associated with more favorable outcomes in men, particularly for anti-CTLA-4 agents. In melanoma, not gender-related factors may influence the anti-tumor immune response evoked by ICIs.
Collapse
Affiliation(s)
- Antonino Grassadonia
- Department of Medical, Oral and Biotechnological Sciences and CeSI-MeT, G. D'Annunzio University, 66100 Chieti, Italy.
| | - Isabella Sperduti
- Department of Bio-Statistics, RCCS Regina Elena National Cancer Institute, 00144 Rome, Italy.
| | - Patrizia Vici
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy.
| | - Laura Iezzi
- Department of Medical, Oral and Biotechnological Sciences and CeSI-MeT, G. D'Annunzio University, 66100 Chieti, Italy.
| | - Davide Brocco
- Department of Medical, Oral and Biotechnological Sciences and CeSI-MeT, G. D'Annunzio University, 66100 Chieti, Italy.
| | - Teresa Gamucci
- Medical Oncology, Sandro Pertini Hospital, 00157 Rome, Italy.
| | - Laura Pizzuti
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy.
| | - Marcello Maugeri-Saccà
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy.
- Scientific Direction, Regina Elena National Cancer Institute, 00144 Rome, Italy.
| | - Paolo Marchetti
- Oncology Unit, Department of Clinical and Molecular Medicine, Medical Oncology, Sapienza University, 00185 Rome, Italy.
| | - Gaetana Cognetti
- Digital library, Knowledge Center "Riccardo Maceratini" and Patient Library, Regina Elena National Cancer Institute, 00144 Rome, Italy.
| | - Michele De Tursi
- Department of Medical, Oral and Biotechnological Sciences and CeSI-MeT, G. D'Annunzio University, 66100 Chieti, Italy.
| | - Clara Natoli
- Department of Medical, Oral and Biotechnological Sciences and CeSI-MeT, G. D'Annunzio University, 66100 Chieti, Italy.
| | - Maddalena Barba
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy.
| | - Nicola Tinari
- Department of Medical, Oral and Biotechnological Sciences and CeSI-MeT, G. D'Annunzio University, 66100 Chieti, Italy.
| |
Collapse
|
3215
|
|
3216
|
Retz M, Bedke J, Bögemann M, Grimm MO, Zimmermann U, Müller L, Leiber C, Teber D, Wirth M, Bolenz C, van Alphen R, De Santis M, Beeker A, Lehmann J, Indorf M, Frank M, Bokemeyer C, Gschwend JE. SWITCH II: Phase III randomized, sequential, open-label study to evaluate the efficacy and safety of sorafenib-pazopanib versus pazopanib-sorafenib in the treatment of advanced or metastatic renal cell carcinoma (AUO AN 33/11). Eur J Cancer 2018; 107:37-45. [PMID: 30529901 DOI: 10.1016/j.ejca.2018.11.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Revised: 10/31/2018] [Accepted: 11/05/2018] [Indexed: 12/22/2022]
Abstract
PURPOSE This trial compared the sequential therapy with the multikinase inhibitor sorafenib (So) followed by pazopanib (Pa) or vice versa in advanced/metastatic renal cell carcinoma (mRCC) patients. METHODS This multicenter, randomized phase 3 study assessed the sequential use of So-Pa versus Pa-So in patients with mRCC without prior systemic therapy. Pts were randomized to So 2 × 400 mg/day followed by Pa 1 × 800 mg/day in case of progression or intolerable toxicity or vice versa. Primary endpoint was total PFS (tPFS), defined as time from randomization to progression, or death during second-line therapy. Key secondary endpoints included overall survival (OS), first-line PFS, disease control rate (DCR) and safety. RESULTS A total of 377 pts were randomized (So-Pa, n = 189; Pa-So, n = 188). Recruitment of a total 544 pts was calculated, but actual accrual rate turned out to be lower than expected. The primary endpoint median tPFS was 8.6 mo (95% CI 7.7-10.2) for So-Pa and 12.9 mo (95% CI 10.8-15.2) for Pa-So with a hazard ratio (HR) of 1.36 (upper limit of one-sided 95% CI 1.68), which exceeded a predefined HR <1.225 as a one-sided 95% confidence interval. Non-inferiority of So-Pa regarding tPFS was not met. Secondary endpoints displayed marked statistical differences in favor of Pa-So in first-line PFS and DCR but not for OS and 2nd-line PFS. Side effect profiles were consistent with known toxicities of the respective multikinase-inhibitor including diarrhea, fatigue, hand-foot skin reaction and hypertension. CONCLUSIONS Non-inferiority of the primary endpoint tPFS could not be demonstrated for So-Pa. The results for first-line PFS and DCR favored the Pa-So sequence. TRIAL REGISTRATION NCT01613846, www.clinicaltrials.gov.
Collapse
Affiliation(s)
- Margitta Retz
- Dept. of Urology, Rechts der Isar Medical Center, Technical University of Munich, Ismaninger Str. 22, 81675 Munich, Germany; AUO Study Group, Germany.
| | - Jens Bedke
- Dept. of Urology, Eberhard Karls University Tübingen, Hoppe-Seyler-Str.3, 72076 Tübingen, Germany.
| | - Martin Bögemann
- Dept. of Urology, Universitätsklinikum Münster, Albert-Schweitzer-Campus 1, 48149 Münster, Germany.
| | - Marc-Oliver Grimm
- Dept. of Urology, Universitätsklinikum Jena, Lessingstr. 1, 07743 Jena, Germany.
| | - Uwe Zimmermann
- Dept. of Urology, Universitätsmedizin Greifswald, Ferdinand-Sauerbruch-Str., 17475 Greifswald, Germany.
| | - Lothar Müller
- Medical Oncology, Onkologie Leer-Emden-Papenburg, Annenestr. 11, 26789 Leer, Germany.
| | - Christian Leiber
- Dept. of Urology, Universitätsklinikum Freiburg, Hugstetter Str. 55, 79106 Freiburg, Germany.
| | - Dogu Teber
- Dept. of Urology, Universitätsklinikum Heidelberg, Im Neuenheimer Feld 110, 69120 Heidelberg, Germany.
| | - Manfred Wirth
- Dept. of Urology, Universitätsklinikum Dresden, Fetscherstr. 74, 01307 Dresden, Germany.
| | - Christian Bolenz
- Dept. of Urology, Universitätsklinikum Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany.
| | - Robbert van Alphen
- Dept. of Oncology, Elisabeth Tweesteden Ziekenhuis, Dr. Deelenlaan 5, 5042 AD Tilburg, Netherlands.
| | - Maria De Santis
- Dept. of Oncology, Kaiser-Franz-Josef-Spital and Department of Urology, Medical University of Vienna, Kundratstr. 3, 1100 Vienna, Austria.
| | - Aart Beeker
- Dept. of Oncology, Spaarne Ziekenhuis, Spaarnepoort 1, 2134 TM Hoofddorp, Netherlands.
| | - Jan Lehmann
- Dept. of Urology, Städtisches Krankenhaus Kiel, Chemnitzstr. 33, 24116 Kiel, Germany; AUO Study Group, Germany.
| | - Martin Indorf
- IOMEDICO AG, Hanferstr. 28, 79108 Freiburg, Germany.
| | - Melanie Frank
- IOMEDICO AG, Hanferstr. 28, 79108 Freiburg, Germany.
| | - Carsten Bokemeyer
- Center for Oncology, II. Medical Clinic and Polyclinic, Universitätsklinikum Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany.
| | - Jürgen E Gschwend
- Dept. of Urology, Rechts der Isar Medical Center, Technical University of Munich, Ismaninger Str. 22, 81675 Munich, Germany; AUO Study Group, Germany.
| |
Collapse
|
3217
|
Affiliation(s)
- Marc-Oliver Grimm
- Urologische Klinik und Poliklinik, Universitätsklinikum Jena, Am Klinikum 1, 07747, Jena, Deutschland.
| |
Collapse
|
3218
|
Hilmi M, Jouinot A, Burns R, Pigneur F, Mounier R, Gondin J, Neuzillet C, Goldwasser F. Body composition and sarcopenia: The next-generation of personalized oncology and pharmacology? Pharmacol Ther 2018; 196:135-159. [PMID: 30521882 DOI: 10.1016/j.pharmthera.2018.12.003] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Body composition has gained increasing attention in oncology in recent years due to fact that sarcopenia has been revealed to be a strong prognostic indicator for survival across multiple stages and cancer types and a predictive factor for toxicity and surgery complications. Accumulating evidence over the last decade has unraveled the "pharmacology" of sarcopenia. Lean body mass may be more relevant to define drug dosing than the "classical" body surface area or flat-fixed dosing in patients with cancer. Since sarcopenia has a major impact on patient survival and quality of life, therapeutic interventions aiming at reducing muscle loss have been developed and are being prospectively evaluated in randomized controlled trials. It is now acknowledged that this supportive care dimension of oncological management is essential to ensure the success of any anticancer treatment. The field of sarcopenia and body composition in cancer is developing quickly, with (i) the newly identified concept of sarcopenic obesity defined as a specific pathophysiological entity, (ii) unsolved issues regarding the best evaluation modalities and cut-off for definition of sarcopenia on imaging, (iii) first results from clinical trials evaluating physical activity, and (iv) emerging body-composition-tailored drug administration schemes. In this context, we propose a comprehensive review providing a panoramic approach of the clinical, pharmacological and therapeutic implications of sarcopenia and body composition in oncology.
Collapse
Affiliation(s)
- Marc Hilmi
- Department of Medical Oncology, CAncer Research for PErsonalized Medicine (CARPEM), Paris Centre Teaching Hospitals, Paris Descartes University, USPC, Paris, France
| | - Anne Jouinot
- Department of Medical Oncology, CAncer Research for PErsonalized Medicine (CARPEM), Paris Centre Teaching Hospitals, Paris Descartes University, USPC, Paris, France
| | - Robert Burns
- Department of Radiology, Henri Mondor University Hospital, Créteil, France
| | - Frédéric Pigneur
- Department of Radiology, Henri Mondor University Hospital, Créteil, France
| | - Rémi Mounier
- Institut NeuroMyoGène (INMG) CNRS 5310 - INSERM U1217 - UCBL, Lyon, France
| | - Julien Gondin
- Institut NeuroMyoGène (INMG) CNRS 5310 - INSERM U1217 - UCBL, Lyon, France
| | - Cindy Neuzillet
- Department of Medical Oncology, Curie Institute, Versailles Saint-Quentin University, Saint-Cloud, France, and GERCOR group, Paris, France.
| | - François Goldwasser
- Department of Medical Oncology, CAncer Research for PErsonalized Medicine (CARPEM), Paris Centre Teaching Hospitals, Paris Descartes University, USPC, Paris, France
| |
Collapse
|
3219
|
Di Nunno V, Gatto L, Fragomeno B, Cubelli M, Nobili E, Romano I, Santoni M, Pisconti S, Montironi R, Massari F. Combination immunotherapy in metastatic renal cell carcinoma. Are we leaving something back? Future Oncol 2018; 14:2997-2999. [DOI: 10.2217/fon-2018-0604] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
| | - Lidia Gatto
- Oncology Unit, SG Moscati Hospital of Taranto, Taranto, Italy
| | | | - Marta Cubelli
- Division of Oncology, S Orsola-Malpighi Hospital, Bologna, Italy
| | | | - Ida Romano
- Division of Oncology, S Orsola-Malpighi Hospital, Bologna, Italy
| | - Matteo Santoni
- Oncology Unit, Macerata Hospital, via Santa Lucia 2, Macerata, Italy
| | | | - Rodolfo Montironi
- Section of Pathological Anatomy, School of Medicine, United Hospital, Polytechnic University of the Marche Region, Ancona, Italy
| | | |
Collapse
|
3220
|
Wang DY, Salem JE, Cohen JV, Chandra S, Menzer C, Ye F, Zhao S, Das S, Beckermann KE, Ha L, Rathmell WK, Ancell KK, Balko JM, Bowman C, Davis EJ, Chism DD, Horn L, Long GV, Carlino MS, Lebrun-Vignes B, Eroglu Z, Hassel JC, Menzies AM, Sosman JA, Sullivan RJ, Moslehi JJ, Johnson DB. Fatal Toxic Effects Associated With Immune Checkpoint Inhibitors: A Systematic Review and Meta-analysis. JAMA Oncol 2018; 4:1721-1728. [PMID: 30242316 PMCID: PMC6440712 DOI: 10.1001/jamaoncol.2018.3923] [Citation(s) in RCA: 1803] [Impact Index Per Article: 257.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Importance Immune checkpoint inhibitors (ICIs) are now a mainstay of cancer treatment. Although rare, fulminant and fatal toxic effects may complicate these otherwise transformative therapies; characterizing these events requires integration of global data. Objective To determine the spectrum, timing, and clinical features of fatal ICI-associated toxic effects. Design, Setting, and Participants We retrospectively queried a World Health Organization (WHO) pharmacovigilance database (Vigilyze) comprising more than 16 000 000 adverse drug reactions, and records from 7 academic centers. We performed a meta-analysis of published trials of anti-programmed death-1/ligand-1 (PD-1/PD-L1) and anti-cytotoxic T lymphocyte antigen-4 (CTLA-4) to evaluate their incidence using data from large academic medical centers, global WHO pharmacovigilance data, and all published ICI clinical trials of patients with cancer treated with ICIs internationally. Exposures Anti-CTLA-4 (ipilimumab or tremelimumab), anti-PD-1 (nivolumab, pembrolizumab), or anti-PD-L1 (atezolizumab, avelumab, durvalumab). Main Outcomes and Measures Timing, spectrum, outcomes, and incidence of ICI-associated toxic effects. Results Internationally, 613 fatal ICI toxic events were reported from 2009 through January 2018 in Vigilyze. The spectrum differed widely between regimens: in a total of 193 anti-CTLA-4 deaths, most were usually from colitis (135 [70%]), whereas anti-PD-1/PD-L1-related fatalities were often from pneumonitis (333 [35%]), hepatitis (115 [22%]), and neurotoxic effects (50 [15%]). Combination PD-1/CTLA-4 deaths were frequently from colitis (32 [37%]) and myocarditis (22 [25%]). Fatal toxic effects typically occurred early after therapy initiation for combination therapy, anti-PD-1, and ipilimumab monotherapy (median 14.5, 40, and 40 days, respectively). Myocarditis had the highest fatality rate (52 [39.7%] of 131 reported cases), whereas endocrine events and colitis had only 2% to 5% reported fatalities; 10% to 17% of other organ-system toxic effects reported had fatal outcomes. Retrospective review of 3545 patients treated with ICIs from 7 academic centers revealed 0.6% fatality rates; cardiac and neurologic events were especially prominent (43%). Median time from symptom onset to death was 32 days. A meta-analysis of 112 trials involving 19 217 patients showed toxicity-related fatality rates of 0.36% (anti-PD-1), 0.38% (anti-PD-L1), 1.08% (anti-CTLA-4), and 1.23% (PD-1/PD-L1 plus CTLA-4). Conclusions and Relevance In the largest evaluation of fatal ICI-associated toxic effects published to date to our knowledge, we observed early onset of death with varied causes and frequencies depending on therapeutic regimen. Clinicians across disciplines should be aware of these uncommon lethal complications.
Collapse
Affiliation(s)
- Daniel Y. Wang
- Vanderbilt Ingram Cancer Center, Department of
Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Joe-Elie Salem
- Vanderbilt Ingram Cancer Center, Department of
Medicine, Vanderbilt University Medical Center, Nashville, Tennessee,Department of Pharmacology, Pharmacovigilance Unit,
AP-HP, Pitié-Salpêtrière Hospital, Paris, France,INSERM, UMR ICAN 1166, Sorbonne Universités, UPMC
Univ Paris 06, Institute of Cardiometabolism and Nutrition (ICAN), Paris, France
| | - Justine V. Cohen
- Department of Medicine, Massachusetts General
Hospital, Boston, Massachusetts
| | - Sunandana Chandra
- Robert H. Lurie Cancer Center, Department of Medicine,
Northwestern University Medical Center, Chicago, Illinois
| | - Christian Menzer
- National Center for Tumor Diseases, Department of
Dermatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Fei Ye
- Vanderbilt Ingram Cancer Center, Department of
Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Shilin Zhao
- Vanderbilt Ingram Cancer Center, Department of
Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Satya Das
- Vanderbilt Ingram Cancer Center, Department of
Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Kathryn E. Beckermann
- Vanderbilt Ingram Cancer Center, Department of
Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Lisa Ha
- Robert H. Lurie Cancer Center, Department of Medicine,
Northwestern University Medical Center, Chicago, Illinois
| | - W. Kimryn Rathmell
- Vanderbilt Ingram Cancer Center, Department of
Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Kristin K. Ancell
- Vanderbilt Ingram Cancer Center, Department of
Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Justin M. Balko
- Vanderbilt Ingram Cancer Center, Department of
Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Caitlin Bowman
- Robert H. Lurie Cancer Center, Department of Medicine,
Northwestern University Medical Center, Chicago, Illinois
| | - Elizabeth J. Davis
- Vanderbilt Ingram Cancer Center, Department of
Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - David D. Chism
- Vanderbilt Ingram Cancer Center, Department of
Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Leora Horn
- Vanderbilt Ingram Cancer Center, Department of
Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Georgina V. Long
- Melanoma Institute of Australia, Sydney, New South
Wales, Australia,University of Sydney, Sydney, New South Wales,
Australia,Royal North Shore Hospital, Sydney, New South Wales,
Australia,Mater Hospital, Sydney, New South Wales,
Australia
| | - Matteo S. Carlino
- Melanoma Institute of Australia, Sydney, New South
Wales, Australia,University of Sydney, Sydney, New South Wales,
Australia,Department of Medical Oncology, Westmeade Hospital,
Sydney, New South Wales, Australia,Department of Medical Oncology, Blacktown Hospital,
Sydney, New South Wales, Australia
| | - Benedicte Lebrun-Vignes
- Department of Pharmacology, Pharmacovigilance Unit,
AP-HP, Pitié-Salpêtrière Hospital, Paris, France,INSERM, UMR ICAN 1166, Sorbonne Universités, UPMC
Univ Paris 06, Institute of Cardiometabolism and Nutrition (ICAN), Paris, France
| | - Zeynep Eroglu
- Department of Cutaneous Oncology, The Moffitt Cancer
Center and Research Institute, Tampa, Florida
| | - Jessica C. Hassel
- National Center for Tumor Diseases, Department of
Dermatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Alexander M. Menzies
- Melanoma Institute of Australia, Sydney, New South
Wales, Australia,University of Sydney, Sydney, New South Wales,
Australia,Department of Medical Oncology, Westmeade Hospital,
Sydney, New South Wales, Australia,Department of Medical Oncology, Blacktown Hospital,
Sydney, New South Wales, Australia
| | - Jeffrey A. Sosman
- Robert H. Lurie Cancer Center, Department of Medicine,
Northwestern University Medical Center, Chicago, Illinois
| | - Ryan J. Sullivan
- Department of Medicine, Massachusetts General
Hospital, Boston, Massachusetts
| | - Javid J. Moslehi
- Vanderbilt Ingram Cancer Center, Department of
Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Douglas B. Johnson
- Vanderbilt Ingram Cancer Center, Department of
Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
3221
|
Wang DY, Salem JE, Cohen JV, Chandra S, Menzer C, Ye F, Zhao S, Das S, Beckermann KE, Ha L, Rathmell WK, Ancell KK, Balko JM, Bowman C, Davis EJ, Chism DD, Horn L, Long GV, Carlino MS, Lebrun-Vignes B, Eroglu Z, Hassel JC, Menzies AM, Sosman JA, Sullivan RJ, Moslehi JJ, Johnson DB. Fatal Toxic Effects Associated With Immune Checkpoint Inhibitors: A Systematic Review and Meta-analysis. JAMA Oncol 2018. [PMID: 30242316 DOI: 10.1001/jamaoncol.2018.3923erratum.in:jamaoncol4(12):1792] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
IMPORTANCE Immune checkpoint inhibitors (ICIs) are now a mainstay of cancer treatment. Although rare, fulminant and fatal toxic effects may complicate these otherwise transformative therapies; characterizing these events requires integration of global data. OBJECTIVE To determine the spectrum, timing, and clinical features of fatal ICI-associated toxic effects. DESIGN, SETTING, AND PARTICIPANTS We retrospectively queried a World Health Organization (WHO) pharmacovigilance database (Vigilyze) comprising more than 16 000 000 adverse drug reactions, and records from 7 academic centers. We performed a meta-analysis of published trials of anti-programmed death-1/ligand-1 (PD-1/PD-L1) and anti-cytotoxic T lymphocyte antigen-4 (CTLA-4) to evaluate their incidence using data from large academic medical centers, global WHO pharmacovigilance data, and all published ICI clinical trials of patients with cancer treated with ICIs internationally. EXPOSURES Anti-CTLA-4 (ipilimumab or tremelimumab), anti-PD-1 (nivolumab, pembrolizumab), or anti-PD-L1 (atezolizumab, avelumab, durvalumab). MAIN OUTCOMES AND MEASURES Timing, spectrum, outcomes, and incidence of ICI-associated toxic effects. RESULTS Internationally, 613 fatal ICI toxic events were reported from 2009 through January 2018 in Vigilyze. The spectrum differed widely between regimens: in a total of 193 anti-CTLA-4 deaths, most were usually from colitis (135 [70%]), whereas anti-PD-1/PD-L1-related fatalities were often from pneumonitis (333 [35%]), hepatitis (115 [22%]), and neurotoxic effects (50 [15%]). Combination PD-1/CTLA-4 deaths were frequently from colitis (32 [37%]) and myocarditis (22 [25%]). Fatal toxic effects typically occurred early after therapy initiation for combination therapy, anti-PD-1, and ipilimumab monotherapy (median 14.5, 40, and 40 days, respectively). Myocarditis had the highest fatality rate (52 [39.7%] of 131 reported cases), whereas endocrine events and colitis had only 2% to 5% reported fatalities; 10% to 17% of other organ-system toxic effects reported had fatal outcomes. Retrospective review of 3545 patients treated with ICIs from 7 academic centers revealed 0.6% fatality rates; cardiac and neurologic events were especially prominent (43%). Median time from symptom onset to death was 32 days. A meta-analysis of 112 trials involving 19 217 patients showed toxicity-related fatality rates of 0.36% (anti-PD-1), 0.38% (anti-PD-L1), 1.08% (anti-CTLA-4), and 1.23% (PD-1/PD-L1 plus CTLA-4). CONCLUSIONS AND RELEVANCE In the largest evaluation of fatal ICI-associated toxic effects published to date to our knowledge, we observed early onset of death with varied causes and frequencies depending on therapeutic regimen. Clinicians across disciplines should be aware of these uncommon lethal complications.
Collapse
Affiliation(s)
- Daniel Y Wang
- Vanderbilt Ingram Cancer Center, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Joe-Elie Salem
- Vanderbilt Ingram Cancer Center, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Pharmacology, Pharmacovigilance Unit, AP-HP, Pitié-Salpêtrière Hospital, Paris, France
- INSERM, UMR ICAN 1166, Sorbonne Universités, UPMC Univ Paris 06, Institute of Cardiometabolism and Nutrition (ICAN), Paris, France
| | - Justine V Cohen
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Sunandana Chandra
- Robert H. Lurie Cancer Center, Department of Medicine, Northwestern University Medical Center, Chicago, Illinois
| | - Christian Menzer
- National Center for Tumor Diseases, Department of Dermatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Fei Ye
- Vanderbilt Ingram Cancer Center, Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Shilin Zhao
- Vanderbilt Ingram Cancer Center, Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Satya Das
- Vanderbilt Ingram Cancer Center, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Kathryn E Beckermann
- Vanderbilt Ingram Cancer Center, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Lisa Ha
- Robert H. Lurie Cancer Center, Department of Medicine, Northwestern University Medical Center, Chicago, Illinois
| | - W Kimryn Rathmell
- Vanderbilt Ingram Cancer Center, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Kristin K Ancell
- Vanderbilt Ingram Cancer Center, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Justin M Balko
- Vanderbilt Ingram Cancer Center, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Caitlin Bowman
- Robert H. Lurie Cancer Center, Department of Medicine, Northwestern University Medical Center, Chicago, Illinois
| | - Elizabeth J Davis
- Vanderbilt Ingram Cancer Center, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - David D Chism
- Vanderbilt Ingram Cancer Center, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Leora Horn
- Vanderbilt Ingram Cancer Center, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Georgina V Long
- Melanoma Institute of Australia, Sydney, New South Wales, Australia
- University of Sydney, Sydney, New South Wales, Australia
- Royal North Shore Hospital, Sydney, New South Wales, Australia
- Mater Hospital, Sydney, New South Wales, Australia
| | - Matteo S Carlino
- Melanoma Institute of Australia, Sydney, New South Wales, Australia
- University of Sydney, Sydney, New South Wales, Australia
- Department of Medical Oncology, Westmeade Hospital, Sydney, New South Wales, Australia
- Department of Medical Oncology, Blacktown Hospital, Sydney, New South Wales, Australia
| | - Benedicte Lebrun-Vignes
- Department of Pharmacology, Pharmacovigilance Unit, AP-HP, Pitié-Salpêtrière Hospital, Paris, France
- INSERM, UMR ICAN 1166, Sorbonne Universités, UPMC Univ Paris 06, Institute of Cardiometabolism and Nutrition (ICAN), Paris, France
| | - Zeynep Eroglu
- Department of Cutaneous Oncology, The Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Jessica C Hassel
- National Center for Tumor Diseases, Department of Dermatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Alexander M Menzies
- Melanoma Institute of Australia, Sydney, New South Wales, Australia
- University of Sydney, Sydney, New South Wales, Australia
- Department of Medical Oncology, Westmeade Hospital, Sydney, New South Wales, Australia
- Department of Medical Oncology, Blacktown Hospital, Sydney, New South Wales, Australia
| | - Jeffrey A Sosman
- Robert H. Lurie Cancer Center, Department of Medicine, Northwestern University Medical Center, Chicago, Illinois
| | - Ryan J Sullivan
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Javid J Moslehi
- Vanderbilt Ingram Cancer Center, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Douglas B Johnson
- Vanderbilt Ingram Cancer Center, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
3222
|
Overview of Microsatellite Instability and Immune Checkpoint Inhibitors in Colorectal Cancer. CURRENT COLORECTAL CANCER REPORTS 2018. [DOI: 10.1007/s11888-018-0413-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
3223
|
Zhang T, Armstrong AJ, George DJ, Huang J. The promise of immunotherapy in genitourinary malignancies. PRECISION CLINICAL MEDICINE 2018; 1:97-101. [PMID: 30687563 PMCID: PMC6333044 DOI: 10.1093/pcmedi/pby018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 11/22/2018] [Indexed: 12/24/2022] Open
Abstract
A broad understanding of the tumor immune landscape has led to a revolution of immune checkpoint inhibitors in the treatment of multiple cancer types. In genitourinary malignancies, immune checkpoint inhibitors have improved outcomes for patients with metastatic renal cell carcinoma and metastatic urothelial carcinoma; however, these treatments have not yet proven broadly beneficial for patients with metastatic prostate cancer. Numerous prospective trials are ongoing to further improve outcomes with immunotherapy combinations and for biomarker development to predict benefit from immune checkpoint inhibition. This perspective article highlights our current immunotherapy approaches in each of the genitourinary malignancies and the ongoing clinical trials that may inform our future treatments in renal, urothelial, and prostate cancers.
Collapse
Affiliation(s)
- Tian Zhang
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC
| | | | - Daniel J George
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC
| | - Jiaoti Huang
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC
| |
Collapse
|
3224
|
Bex A, Albiges L, Staehler M, Bensalah K, Giles RH, Dabestani S, Hofmann F, Hora M, Kuczyk MA, Lam TB, Marconi L, Merseburger AS, Fernández-Pello S, Tahbaz R, Abu-Ghanem Y, Volpe A, Ljungberg B, Escudier B, Powles T. A Joint Statement from the European Association of Urology Renal Cell Cancer Guidelines Panel and the International Kidney Cancer Coalition: The Rejection of Ipilimumab and Nivolumab for Renal Cancer by the Committee for Medicinal Products for Human Use Does not Change Evidence-based Guideline Recommendations. Eur Urol 2018; 74:849-851. [PMID: 30201510 DOI: 10.1016/j.eururo.2018.08.029] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Accepted: 08/20/2018] [Indexed: 10/28/2022]
Affiliation(s)
- Axel Bex
- Department of Urology, The Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Laurence Albiges
- Department of Cancer Medicine, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Michael Staehler
- Department of Urology, Ludwig-Maximilians University, Munich, Germany
| | - Karim Bensalah
- Department of Urology, University of Rennes, Rennes, France
| | - Rachel H Giles
- International Kidney Cancer Coalition (IKCC), Duivendrecht, The Netherlands; Department of Nephrology and Hypertension, Regenerative Medicine Center, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Saeed Dabestani
- Department of Clinical Sciences Lund, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Fabian Hofmann
- Department of Urology, Sunderby Hospital, Sunderby, Sweden
| | - Milan Hora
- Department of Urology, Faculty Hospital and Faculty of Medicine in Pilsen, Charles University in Prague, Prague, Czech Republic
| | - Markus A Kuczyk
- Department of Urology and Urologic Oncology, Hannover Medical School, Hannover, Germany
| | - Thomas B Lam
- Department of Urology, Aberdeen Royal Infirmary, Aberdeen, UK; Academic Urology Unit, University of Aberdeen, Aberdeen, UK
| | - Lorenzo Marconi
- Department of Urology, Coimbra University Hospital, Coimbra, Portugal
| | - Axel S Merseburger
- Department of Urology, University Hospital Schleswig-Holstein, Lübeck, Germany
| | | | - Rana Tahbaz
- Department of Urology, Elbe Kliniken Stade, Stade, Germany
| | - Yasmin Abu-Ghanem
- Department of Urology, Chaim Sheba Medical Center, Tel-Hashomer, Ramat-Gan, Israel
| | - Alessandro Volpe
- Division of Urology, Maggiore della Carita' Hospital, University of Eastern Piedmont, Novara, Italy
| | - Börje Ljungberg
- Department of Surgical and Perioperative Sciences, Urology and Andrology, Umeå University, Umeå, Sweden
| | | | - Thomas Powles
- The Royal Free NHS Trust and Barts Cancer Institute, Queen Mary University of London, London, UK.
| |
Collapse
|
3225
|
Herrera AF. Where does PD-1 blockade fit in HL therapy? HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2018; 2018:213-220. [PMID: 30504313 PMCID: PMC6246012 DOI: 10.1182/asheducation-2018.1.213] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Genetic alterations of the PD-L1/PD-L2 locus on chromosome 9p24.1 are a defining biological feature of classical Hodgkin lymphoma (HL). The resulting programmed death-ligand 1 (PD-L1) expression on Hodgkin Reed-Sternberg cells as well as the PD-L1 expressed in the HL microenvironment result in an ineffective host antitumor immune response and make HL a ripe target for programmed cell death-1 (PD-1) blockade. Anti-PD-1 antibody monotherapy has been effective and well tolerated in patients with relapsed or refractory (rel/ref) HL, with the majority of patients experiencing an objective response (approximately two-thirds of patients) and a median duration of response of 16.6 months in the study with the longest follow-up. Based on these data, nivolumab and pembrolizumab were approved by the US Food and Drug Administration (FDA) for the treatment of advanced rel/ref HL. Evidence has emerged that patients with HL benefit from continued PD-1 blockade beyond disease progression according to traditionally defined response criteria, and that the addition of, or switch to, chemotherapy after anti-PD-1 antibody failure can potentially re-induce clinical response. Subsequent studies have evaluated novel anti-PD-1-based combination regimens as well as the use of anti-PD-1 antibody therapy earlier in the course of a HL patient's therapy, including first salvage therapy for rel/ref disease (eg, nivolumab plus brentuximab vedotin) and even first-line treatment (eg, nivolumab added to doxorubicin, vinblastine, dacarbazine chemotherapy). The current role of PD-1 blockade in HL is as monotherapy in patients with advanced rel/ref disease, but the results of ongoing studies and the evolving treatment landscape in HL will determine the role of PD-1 blockade in the future.
Collapse
Affiliation(s)
- Alex F Herrera
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA
| |
Collapse
|
3226
|
Williams P, Basu S, Garcia-Manero G, Hourigan CS, Oetjen KA, Cortes JE, Ravandi F, Jabbour EJ, Al-Hamal Z, Konopleva M, Ning J, Xiao L, Hidalgo Lopez J, Kornblau SM, Andreeff M, Flores W, Bueso-Ramos C, Blando J, Galera P, Calvo KR, Al-Atrash G, Allison JP, Kantarjian HM, Sharma P, Daver NG. The distribution of T-cell subsets and the expression of immune checkpoint receptors and ligands in patients with newly diagnosed and relapsed acute myeloid leukemia. Cancer 2018; 125:1470-1481. [PMID: 30500073 PMCID: PMC6467779 DOI: 10.1002/cncr.31896] [Citation(s) in RCA: 243] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 09/07/2018] [Accepted: 10/09/2018] [Indexed: 12/26/2022]
Abstract
Background Phenotypic characterization of immune cells in the bone marrow (BM) of patients with acute myeloid leukemia (AML) is lacking. Methods T‐cell infiltration was quantified on BM biopsies from 13 patients with AML, and flow cytometry was performed on BM aspirates (BMAs) from 107 patients with AML who received treatment at The University of Texas MD Anderson Cancer Center. The authors evaluated the expression of inhibitory receptors (programmed cell death protein 1 [PD1], cytotoxic T‐lymphocyte antigen 4 [CTLA4], lymphocyte‐activation gene 3 [LAG3], T‐cell immunoglobulin and mucin‐domain containing‐3 [TIM3]) and stimulatory receptors (glucocorticoid‐induced tumor necrosis factor receptor‐related protein [GITR], OX40, 41BB [a type 2 transmembrane glycoprotein receptor], inducible T‐cell costimulatory [ICOS]) on T‐cell subsets and the expression of their ligands (41BBL, B7‐1, B7‐2, ICOSL, PD‐L1, PD‐L2, and OX40L) on AML blasts. Expression of these markers was correlated with patient age, karyotype, baseline next‐generation sequencing for 28 myeloid‐associated genes (including P53), and DNA methylation proteins (DNA methyltransferase 3α, isocitrate dehydrogenase 1[IDH1], IDH2, Tet methylcytosine dioxygenase 2 [TET2], and Fms‐related tyrosine kinase 3 [FLT3]). Results On histochemistry evaluation, the T‐cell population in BM appeared to be preserved in patients who had AML compared with healthy donors. The proportion of T‐regulatory cells (Tregs) in BMAs was higher in patients with AML than in healthy donors. PD1‐positive/OX40‐positive T cells were more frequent in AML BMAs, and a higher frequency of PD1‐positive/cluster of differentiation 8 (CD8)‐positive T cells coexpressed TIM3 or LAG3. PD1‐positive/CD8‐positive T cells were more frequent in BMAs from patients who had multiply relapsed AML than in BMAs from those who had first relapsed or newly diagnosed AML. Blasts in BMAs from patients who had TP53‐mutated AML were more frequently positive for PD‐L1. Conclusions The preserved T‐cell population, the increased frequency of regulatory T cells, and the expression of targetable immune receptors in AML BMAs suggest a role for T‐cell–harnessing therapies in AML. T‐cell subsets are preserved in the bone marrow of patients with acute myeloid leukemia. The expression of targetable immune checkpoints by T cells suggests that therapies harnessing T cells may benefit these patients.
Collapse
Affiliation(s)
- Patrick Williams
- Department of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sreyashi Basu
- Immunotherapy Platform, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Christopher S Hourigan
- Laboratory of Myeloid Malignancies, Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Karolyn A Oetjen
- Laboratory of Myeloid Malignancies, Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Jorge E Cortes
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Farhad Ravandi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Elias J Jabbour
- Department of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Zainab Al-Hamal
- Immunotherapy Platform, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Marina Konopleva
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jing Ning
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lianchun Xiao
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Juliana Hidalgo Lopez
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Steve M Kornblau
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael Andreeff
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Wilmer Flores
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Carlos Bueso-Ramos
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jorge Blando
- Immunotherapy Platform, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Pallavi Galera
- Department of Laboratory Medicine, National Institutes of Health Clinical Center, Bethesda, Maryland
| | - Katherine R Calvo
- Department of Laboratory Medicine, National Institutes of Health Clinical Center, Bethesda, Maryland
| | - Gheath Al-Atrash
- Department of Stem Cell Transplant and Cell Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - James P Allison
- Immunotherapy Platform, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hagop M Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Padmanee Sharma
- Immunotherapy Platform, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Naval G Daver
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
3227
|
Furubayashi N, Negishi T, Uozumi T, Takamatsu D, Shiraishi K, Hirose D, Nakamura M. Isolated adrenocorticotropic hormone deficiency potentially induced by nivolumab following pseudo-progression in clear cell renal cell carcinoma: A case report. Mol Clin Oncol 2018; 10:304-308. [PMID: 30680212 DOI: 10.3892/mco.2018.1781] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 11/27/2018] [Indexed: 01/05/2023] Open
Abstract
Nivolumab is a monoclonal immunoglobulin G antibody blocking programmed death receptor-1 (PD-1) that promotes the restoration of the natural T-cell-mediated immune response against cancer cells; however, it also causes a number of autoimmune-related adverse events (irAEs) that often involve the endocrine system. The present report describes a 71-year-old man with clear cell renal cell carcinoma metastasis in the lung. Following the 14th course of nivolumab therapy, the patient complained of general malaise, loss of appetite and mild consciousness disturbance. Laboratory tests revealed a severely elevated eosinophil ratio (26.2%) and low sodium value (122 mmol/l). Endocrine system tests revealed that the patient's adrenocorticotropic hormone (ACTH; 4.5 pg/ml) and cortisol (0.1 µg/dl) levels were lower than normal, while those of other pituitary hormones were higher than normal. This case was therefore diagnosed as isolated ACTH deficiency induced by nivolumab. Magnetic resonance imaging (MRI) showed normal pituitary glands. Hydrocortisone replacement therapy improved the clinical symptoms early and enabled the patient to restart nivolumab therapy. Isolated ACTH deficiency due to nivolumab, a PD-1 immune checkpoint inhibitor antibody, is a rare occurrence. This report may be useful for avoiding delays in the diagnosis and treatment of this life-threatening irAE even if no pituitary abnormalities are identified via MRI.
Collapse
Affiliation(s)
- Nobuki Furubayashi
- Department of Urology, National Kyushu Cancer Center, Fukuoka 811-1395, Japan
| | - Takahito Negishi
- Department of Urology, National Kyushu Cancer Center, Fukuoka 811-1395, Japan
| | - Tomoharu Uozumi
- Department of Urology, National Kyushu Cancer Center, Fukuoka 811-1395, Japan
| | - Dai Takamatsu
- Department of Urology, National Kyushu Cancer Center, Fukuoka 811-1395, Japan
| | - Koichi Shiraishi
- Department of Urology, National Kyushu Cancer Center, Fukuoka 811-1395, Japan
| | - Daisuke Hirose
- Department of Urology, National Kyushu Cancer Center, Fukuoka 811-1395, Japan
| | - Motonobu Nakamura
- Department of Urology, National Kyushu Cancer Center, Fukuoka 811-1395, Japan
| |
Collapse
|
3228
|
Conway JR, Kofman E, Mo SS, Elmarakeby H, Van Allen E. Genomics of response to immune checkpoint therapies for cancer: implications for precision medicine. Genome Med 2018; 10:93. [PMID: 30497521 PMCID: PMC6264032 DOI: 10.1186/s13073-018-0605-7] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Immune checkpoint blockade (ICB) therapies, which potentiate the body's natural immune response against tumor cells, have shown immense promise in the treatment of various cancers. Currently, tumor mutational burden (TMB) and programmed death ligand 1 (PD-L1) expression are the primary biomarkers evaluated for clinical management of cancer patients across histologies. However, the wide range of responses has demonstrated that the specific molecular and genetic characteristics of each patient's tumor and immune system must be considered to maximize treatment efficacy. Here, we review the various biological pathways and emerging biomarkers implicated in response to PD-(L)1 and cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) therapies, including oncogenic signaling pathways, human leukocyte antigen (HLA) variability, mutation and neoantigen burden, microbiome composition, endogenous retroviruses (ERV), and deficiencies in chromatin remodeling and DNA damage repair (DDR) machinery. We also discuss several mechanisms that have been observed to confer resistance to ICB, such as loss of phosphatase and tensin homolog (PTEN), loss of major histocompatibility complex (MHC) I/II expression, and activation of the indoleamine 2,3-dioxygenase 1 (IDO1) and transforming growth factor beta (TGFβ) pathways. Clinical trials testing the combination of PD-(L)1 or CTLA-4 blockade with molecular mediators of these pathways are becoming more common and may hold promise for improving treatment efficacy and response. Ultimately, some of the genes and molecular mechanisms highlighted in this review may serve as novel biological targets or therapeutic vulnerabilities to improve clinical outcomes in patients.
Collapse
Affiliation(s)
- Jake R Conway
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, 02142, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, 02215, USA
| | - Eric Kofman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, 02142, USA
| | - Shirley S Mo
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, 02142, USA
| | - Haitham Elmarakeby
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, 02142, USA
- Department of System and Computer Engineering, Al-Azhar University, Cairo, 11751, Egypt
| | - Eliezer Van Allen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA.
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, 02142, USA.
| |
Collapse
|
3229
|
Hokland P, Hokland M, Cotter F. The Nobel Prize for Medicine awarded for cancer therapy by inhibition of negative immune regulation. Br J Haematol 2018; 183:698-700. [PMID: 30488426 DOI: 10.1111/bjh.15694] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Peter Hokland
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | | | - Finbarr Cotter
- Centre for Haematology-Oncology, Barts Cancer Institute, Barts and London School of Medicine and Dentistry, London, UK
| |
Collapse
|
3230
|
Santoni M, Conti A, Buti S, Bersanelli M, Foghini L, Piva F, Giulietti M, Lusuardi L, Battelli N. Risk of fatigue in cancer patients treated with anti programmed cell death-1/anti programmed cell death ligand-1 agents: a systematic review and meta-analysis. Immunotherapy 2018; 10:1303-1313. [PMID: 30474475 DOI: 10.2217/imt-2018-0067] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM We aimed to assess the incidence and relative risk (RR) of fatigue in cancer patients treated with anti programmed cell death-1 (PD-1) and anti programmed cell death ligand-1 (PD-L1) agents. PATIENTS & METHODS Eligible studies were selected according to Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement. Incidence, RR and 95% CIs were calculated using random or fixed-effects models. RESULTS Thirty-eight studies were included in this analysis, with a total of 11,719 patients. The incidences were 23.4 and 2.1% for all- and high-grade fatigue, respectively. The highest incidence of high-grade fatigue was reported by the combination of nivolumab and ipilimumab. Overall RR of high-grade fatigue with anti-PD-1/PD-L1 compared with chemotherapy or targeted therapy was 0.48. CONCLUSION Treatment with anti-PD-1/PD-L1 agents correlates with lower incidence and RR of fatigue compared with standard therapies.
Collapse
Affiliation(s)
- Matteo Santoni
- Oncology Unit, Macerata Hospital, via Santa Lucia 2, 62100, Macerata, Italy
| | - Alessandro Conti
- Azienda Ospedaliera dell'Alto Adige, Bressanone/Brixen Hospital, Via Dante, 51, 39042, Italy
| | - Sebastiano Buti
- University Hospital of Parma, Via Gramsci 14 - 43126, Parma, Italy
| | | | - Laura Foghini
- Oncology Unit, Macerata Hospital, via Santa Lucia 2, 62100, Macerata, Italy
| | - Francesco Piva
- Department of Specialistic Clinical & Odontostomatological Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Matteo Giulietti
- Department of Specialistic Clinical & Odontostomatological Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Lukas Lusuardi
- Department of Urology & Andrology, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Nicola Battelli
- Oncology Unit, Macerata Hospital, via Santa Lucia 2, 62100, Macerata, Italy
| |
Collapse
|
3231
|
D'Avella C, Abbosh P, Pal SK, Geynisman DM. Mutations in renal cell carcinoma. Urol Oncol 2018; 38:763-773. [PMID: 30478013 DOI: 10.1016/j.urolonc.2018.10.027] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 10/19/2018] [Accepted: 10/29/2018] [Indexed: 12/14/2022]
Abstract
Renal cell carcinoma (RCC) is a commonly diagnosed and histologically diverse urologic malignancy. Clear cell RCC (ccRCC) is by far the most common, followed by the papillary and chromophobe subtypes. Sarcomatoid differentiation is a morphologic change that can be seen in all subtypes that typically portends a poor prognosis. In the past, treatment options for RCC were limited to cytokine-based therapy with a high-toxicity profile and low response rate. An increased understanding of the molecular basis of RCC has led to substantial improvement in treatment options in the form of targeted therapy and immunotherapy. A significant early discovery in RCC was frequent inactivation of the Von Hippel Lindau gene in ccRCC, which ultimately led to the development of vascular endothelial growth factor and mammalian target of rapamycin inhibitors. Further genomic sequencing of ccRCC tumors has identified other common mutations including BAP-1, PBRM1, SETD2, and PIK3CA. Many recent studies have explored how these mutations can affect prognosis and response to treatment. Likewise, papillary RCC has also been studied at the molecular level, which has shown a high level of mutations in the MET gene; early clinical data suggest the utility of MET targeted therapy. Finally, regarding the rarer sarcomatoid tumors, mutations in TP53 and NF2 may be important to their development. As we continue to learn more about what drives RCC at the molecular level, treatment options for RCC patients are diversifying.
Collapse
Affiliation(s)
| | - Phillip Abbosh
- Molecular Therapeutics, Fox Chase Cancer Center, Temple Health, Philadelphia, PA; Department of Urology, Einstein Medical Center, Philadelphia, PA
| | - Sumanta K Pal
- Department of Medical Oncology & Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Daniel M Geynisman
- Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA.
| |
Collapse
|
3232
|
Wu B, Zhang Q, Sun J. Cost-effectiveness of nivolumab plus ipilimumab as first-line therapy in advanced renal-cell carcinoma. J Immunother Cancer 2018; 6:124. [PMID: 30458884 PMCID: PMC6247499 DOI: 10.1186/s40425-018-0440-9] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 10/31/2018] [Indexed: 12/18/2022] Open
Abstract
Background Nivolumab plus ipilimumab improves overall survival and is associated with less toxicity compared with sunitinib in the first-line setting of advanced renal-cell carcinoma (RCC). The current study aimed to assess the cost-effectiveness of nivolumab plus ipilimumab for first-line treatment of advanced RCC from the payer perspectives high- and middle-income regions. Methods A decision-analytic model was constructed to evaluate the health and economic outcomes of first-line sunitinib and nivolumab plus ipilimumab treatment associated with advanced RCC. The clinical and utility data were obtained from published reports. The cost data were acquired for the payer perspectives of the United States (US), United Kingdom (UK), and China. Sensitivity analyses were performed to test the uncertainties of the results. Quality-adjusted life-years (QALYs) and incremental cost-effectiveness ratios (ICERs) were used. Results Nivolumab plus ipilimumab gained 0.70–0.76 QALYs compared with sunitinib. Our analysis determined the following ICERs for nivolumab plus ipilimumab over sunitinib in first-line advanced RCC treatment: US $ 85,506 /QALY; UK $ 126,499/QALY; and China $ 4682/QALY. Sensitivity analyses found the model outputs to be most affected for body weight and for the prices of nivolumab, sunitinib and ipilimumab. Conclusions Nivolumab plus ipilimumab as first-line treatment could gain more health benefits for advanced RCC in comparison with standard sunitinib, which is considered to be cost-effective in the US and China but not in the UK. Electronic supplementary material The online version of this article (10.1186/s40425-018-0440-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Bin Wu
- Medical Decision and Economic Group, Department of Pharmacy, South Campus, Ren Ji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Qiang Zhang
- Department of Clinical Oncology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jie Sun
- Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China.
| |
Collapse
|
3233
|
Abstract
Renal cell cancer (RCC) (epithelial carcinoma of the kidney) represents 2%-4% of newly diagnosed adult tumors. Over the past 2 decades, RCC has been better characterized clinically and molecularly. It is a heterogeneous disease, with multiple subtypes, each with characteristic histology, genetics, molecular profiles, and biologic behavior. Tremendous heterogeneity has been identified with many distinct subtypes characterized. There are clinical questions to be addressed at every stage of this disease, and new targets being identified for therapeutic development. The unique characteristics of the clinical presentations of RCC have led to both questions and opportunities for improvement in management. Advances in targeted drug development and understanding of immunologic control of RCC are leading to a number of new clinical trials and regimens for advanced disease, with the goal of achieving long-term disease-free survival, as has been achieved in a proportion of such patients historically. RCC management is a promising area of ongoing clinical investigation.
Collapse
|
3234
|
Gonzalez-Cao M, Martinez-Picado J, Karachaliou N, Rosell R, Meyerhans A. Cancer immunotherapy of patients with HIV infection. Clin Transl Oncol 2018; 21:713-720. [PMID: 30446984 DOI: 10.1007/s12094-018-1981-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 11/07/2018] [Indexed: 12/16/2022]
Abstract
Cancer immunotherapy with antibodies against immune checkpoints has made impressive advances in the last several years. The most relevant drugs target programmed cell death 1 (PD-1) expressed on T cells or its ligand, the programmed cell death ligand 1 (PD-L1), expressed on cancer cells, and cytotoxic T lymphocyte-associated protein 4 (CTLA-4). Unfortunately, cancer patients with HIV infection are usually excluded from cancer clinical trials, because there are concerns about the safety and the anti-tumoral activity of these novel therapies in patients with HIV infection. Several retrospective studies and some case reports now support the notion that antibodies against immune checkpoints are safe and active in cancer patients with HIV infection, but prospective data in these patients are lacking. In addition, signs of antiviral activity with increase in CD4 T cell counts, plasma viremia reduction or decrease in the viral reservoir have been reported in some of the patients treated, although no patient achieved a complete clearance of the viral reservoir. Here we briefly summarize all clinical cases reported in the literature, as well as ongoing clinical trials testing novel immunotherapy drugs in cancer patients with HIV infection.
Collapse
Affiliation(s)
- M Gonzalez-Cao
- Dr. Rosell Oncology Institute (IOR), Dexeus University Hospital, Quironsalud Group, C/Sabino Arana, 5, 08028, Barcelona, Spain.
| | - J Martinez-Picado
- AIDS Research Institute IrsiCaixa, Badalona, Spain.,University of Vic-Central University of Catalonia (UVic-UCC), Vic, Spain.,Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - N Karachaliou
- Dr. Rosell Oncology Institute (IOR), Sagrat Cor University Hospital, Quironsalud Group, Barcelona, Spain
| | - R Rosell
- Dr. Rosell Oncology Institute (IOR), Dexeus University Hospital, Quironsalud Group, C/Sabino Arana, 5, 08028, Barcelona, Spain.,Dr. Rosell Oncology Institute (IOR), Sagrat Cor University Hospital, Quironsalud Group, Barcelona, Spain.,Catalan Institute of Oncology, Germans Trias i Pujol University Hospital, Badalona, Spain
| | - A Meyerhans
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain.,Infection Biology Laboratory, Department of Experimental and Health Sciences (DCEXS), Universitat Pompeu Fabra, Barcelona, Spain
| |
Collapse
|
3235
|
Nie RC, Chen FP, Yuan SQ, Luo YS, Chen S, Chen YM, Chen XJ, Chen YB, Li YF, Zhou ZW. Evaluation of objective response, disease control and progression-free survival as surrogate end-points for overall survival in anti-programmed death-1 and anti-programmed death ligand 1 trials. Eur J Cancer 2018; 106:1-11. [PMID: 30453169 DOI: 10.1016/j.ejca.2018.10.011] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 10/09/2018] [Accepted: 10/10/2018] [Indexed: 12/31/2022]
Abstract
BACKGROUND We aimed to assess whether the Response Evaluation Criteria in Solid Tumors (RECIST) criteria-based objective response rate (ORR), disease control rate (DCR) and progression-free survival (PFS) could be valid surrogate end-points for overall survival (OS) in anti-programmed death-1 (PD-1)/programmed death ligand 1 (PD-L1) trials. METHODS We systematically reviewed phase 2 and phase 3 trials of anti-PD-1/PD-L1 drug trials of advanced or recurrent solid tumours that reported OS and at least one of the RECIST criteria-based end-points. We used Spearman rank correlation to evaluate the strength of the association between these end-points and OS and a linear regression model, weighted by the sample size, to assess the association between the treatment effect on these end-points and OS. We also performed sensitivity analyses and a leave-one-out cross-validation approach to evaluate the robustness of our findings. RESULTS Forty-three qualifying trails comprising 15,088 patients were eligible. PFS showed good correlation with OS (squared Spearman rank correlation coefficient [rs2] = 0.54; P < 0.001), while ORR and DCR illustrated moderate association with OS (rs2 = 0.29 and 0.28, respectively; both P < 0.001). The correlation was moderate between the treatment effects on PFS and OS (coefficient of determination [R2] = 0.37, P < 0.001) and poor among ORR, DCR and OS (R2 = 0.10 and 0.08, respectively); these were confirmed by sensitivity analyses (all R2 < 0.75) and the leave-one-out cross-validation approach. CONCLUSIONS No RECIST criteria-based end-points could be a valid surrogate for OS. At present, we proposed to set OS as the primary end-point in anti-PD-1/PD-L1 drug trials of advanced or recurrent solid tumours.
Collapse
Affiliation(s)
- Run-Cong Nie
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Fo-Ping Chen
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| | - Shu-Qiang Yuan
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Ying-Shan Luo
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| | - Shi Chen
- Department of Gastric Surgery, The 6th Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yong-Ming Chen
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Xiao-Jiang Chen
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Ying-Bo Chen
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yuan-Fang Li
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.
| | - Zhi-Wei Zhou
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.
| |
Collapse
|
3236
|
Ayers M, Nebozhyn M, Cristescu R, McClanahan TK, Perini R, Rubin E, Cheng JD, Kaufman DR, Loboda A. Molecular Profiling of Cohorts of Tumor Samples to Guide Clinical Development of Pembrolizumab as Monotherapy. Clin Cancer Res 2018; 25:1564-1573. [PMID: 30442684 DOI: 10.1158/1078-0432.ccr-18-1316] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 06/29/2018] [Accepted: 08/16/2018] [Indexed: 11/16/2022]
Abstract
PURPOSE Molecular profiling of large databases of human tumor gene expression profiles offers novel opportunities for informing decisions in clinical development programs. EXPERIMENTAL DESIGN Gene expression profile of programmed death ligand 1 (PD-L1) was explored in a dataset of 16,000 samples, including approximately 4,000 metastatic tumors, across >25 tumor types prevalent in the United States, looking for new indications for the programmed death 1 (PD-1) inhibitor pembrolizumab. PD-L1 expression was highly concordant with several genomic signatures indicative of immune-inflamed tumor microenvironment. Prevalence of activated immune-inflamed tumors across all tumor types was explored and used to rank tumor types for potential response to pembrolizumab monotherapy. RESULTS The analysis yielded 3 tiers of indications in which high levels of PD-L1 and immune-inflamed signatures were found in up to 40% to 60%, 20% to 40%, and 0% to 20% of tumors. Tier 1 contained novel indications known at the time of analysis to be responsive to PD-1 checkpoint blockade in the clinic (such as melanoma and non-small cell lung cancer), as well as indications not studied in the clinic previously, including microsatellite instability-high colorectal, head and neck, bladder, and triple-negative breast cancers. Complementary analysis of an Asian/Pacific cancer dataset (gastric cancer) revealed high prevalence of immune-inflamed tumors in gastric cancer. These data contributed to prioritization of these indications for clinical development of pembrolizumab as monotherapy. CONCLUSIONS Data highlight the value of molecular profiling in identifying populations with high unmet needs with potentially favorable response characteristics and accelerating development of novel therapies for these patients.See related commentary by Mansfield and Jen, p. 1443.
Collapse
Affiliation(s)
- Mark Ayers
- Merck & Co., Inc., Kenilworth, New Jersey
| | | | | | | | | | - Eric Rubin
- Merck & Co., Inc., Kenilworth, New Jersey
| | | | | | | |
Collapse
|
3237
|
Agarwal N, Nussenzveig R, Pal SK. Biomarkers in renal-cell carcinoma: building on clinical paradigms. Lancet Oncol 2018; 19:1560-1561. [PMID: 30416074 DOI: 10.1016/s1470-2045(18)30674-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 08/31/2018] [Indexed: 11/18/2022]
Affiliation(s)
- Neeraj Agarwal
- Huntsman Cancer Institute at the University of Utah, Salt Lake City, UT 84112, USA.
| | - Roberto Nussenzveig
- Huntsman Cancer Institute at the University of Utah, Salt Lake City, UT 84112, USA
| | - Sumanta K Pal
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| |
Collapse
|
3238
|
Yentz S, Smith D. Indoleamine 2,3-Dioxygenase (IDO) Inhibition as a Strategy to Augment Cancer Immunotherapy. BioDrugs 2018; 32:311-317. [PMID: 29980987 DOI: 10.1007/s40259-018-0291-4] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Indoleamine 2,3-dioxygenase (IDO) is an enzyme of interest in immuno-oncology because of the immunosuppressive effects that result from its role in tryptophan catabolism. IDO is upregulated in malignancy and is associated with poor prognosis in multiple cancer types. IDO inhibitors have been developed to target IDO, both directly and indirectly. Pre-clinical data have shown combined IDO and checkpoint inhibition to be an efficacious strategy for tumor control. Clinical trials of IDO inhibitors with chemotherapy or immunotherapy are currently underway. This review describes the function of IDO and its inhibitors and summarizes the efficacy and toxicity data from recent clinical trials with these drugs.
Collapse
Affiliation(s)
- Sarah Yentz
- Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan Health System, 1500 E. Medical Center Drive, C369 Med Inn Building, SPC 5848, Ann Arbor, MI, 48109, USA.
| | - David Smith
- Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan Health System, 1500 E. Medical Center Drive, C369 Med Inn Building, SPC 5848, Ann Arbor, MI, 48109, USA
| |
Collapse
|
3239
|
Hong B, Cai L, Wang J, Liu S, Zhou J, Ma K, Zhang J, Zhou B, Peng X, Zhang N, Gong K. Differential Expression of PD-L1 Between Sporadic and VHL-Associated Hereditary Clear-Cell Renal Cell Carcinoma and Its Correlation With Clinicopathological Features. Clin Genitourin Cancer 2018; 17:97-104.e1. [PMID: 30522901 DOI: 10.1016/j.clgc.2018.11.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 10/30/2018] [Accepted: 11/04/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND Programmed death ligand-1 (PD-L1) is a potential predictive biomarker for immunotherapy in several malignancies. However, the expression level and clinical significance of PD-L1 in von Hippel-Lindau (VHL)-associated hereditary clear-cell renal cell carcinoma (ccRCC) remain unclear. PATIENTS AND METHODS Surgical specimens were recruited from 129 patients with sporadic ccRCC and 26 patients with VHL-associated hereditary ccRCC. The PD-L1 expression level was assessed using immunohistochemistry. Correlations between PD-L1 expression and clinicopathological features were analyzed. RESULTS In sporadic ccRCC, the positive expression rate of PD-L1 was 47.3% (61/129). Positive PD-L1 expression was correlated with advanced tumor T stage (P = .011), higher Fuhrman nuclear grade (P = .022), poor disease-free survival (P = .037), and sex (P = .025). In the VHL-associated hereditary ccRCC, positive PD-L1 expression rate was 34.6% (9/26), lower than that in sporadic ccRCC. Positive PD-L1 was correlated with higher Fuhrman nuclear grade (P = .008), but not with sex, age, tumor stage, or the onset age of VHL-associated tumors. CONCLUSION Positive PD-L1 expression was correlated with the aggressive clinicopathological features in sporadic and VHL-associated hereditary ccRCC. Whether PD-L1 expression level in ccRCC is related to the effectiveness of programmed death-1/PD-L1 checkpoint inhibitor immunotherapy needs to be further investigated.
Collapse
Affiliation(s)
- Baoan Hong
- Department of Urology, Peking University First Hospital, Beijing, China; Institute of Urology, Peking University, Beijing, China; National Urological Cancer Center, Beijing, China
| | - Lin Cai
- Department of Urology, Peking University First Hospital, Beijing, China; Institute of Urology, Peking University, Beijing, China; National Urological Cancer Center, Beijing, China
| | - Jiangyi Wang
- Department of Urology, Peking University First Hospital, Beijing, China; Institute of Urology, Peking University, Beijing, China; National Urological Cancer Center, Beijing, China
| | - Shengjie Liu
- Department of Urology, Peking University First Hospital, Beijing, China; Institute of Urology, Peking University, Beijing, China; National Urological Cancer Center, Beijing, China
| | - Jingcheng Zhou
- Department of Urology, Peking University First Hospital, Beijing, China; Institute of Urology, Peking University, Beijing, China; National Urological Cancer Center, Beijing, China
| | - Kaifang Ma
- Department of Urology, Peking University First Hospital, Beijing, China; Institute of Urology, Peking University, Beijing, China; National Urological Cancer Center, Beijing, China
| | - Jiufeng Zhang
- Department of Urology, Peking University First Hospital, Beijing, China; Institute of Urology, Peking University, Beijing, China; National Urological Cancer Center, Beijing, China
| | - Bowen Zhou
- Department of Urology, Peking University First Hospital, Beijing, China; Institute of Urology, Peking University, Beijing, China; National Urological Cancer Center, Beijing, China
| | - Xiang Peng
- Department of Urology, Peking University First Hospital, Beijing, China; Institute of Urology, Peking University, Beijing, China; National Urological Cancer Center, Beijing, China
| | - Ning Zhang
- Department of Urology, Beijing Cancer Hospital, Beijing, China.
| | - Kan Gong
- Department of Urology, Peking University First Hospital, Beijing, China; Institute of Urology, Peking University, Beijing, China; National Urological Cancer Center, Beijing, China.
| |
Collapse
|
3240
|
Abstract
PURPOSE OF REVIEW Immune checkpoint blockade targeting PD-1 and PD-L1 improves immune recognition of tumor cells but had only modest success in gynecological cancers as monotherapy. Growing focus has been placed on combination immunotherapy strategies to overcome this resistance, and this review serves to discuss some of the most promising studies in gynecological cancers. RECENT FINDINGS PD-1- and PD-L1-targeting antibodies are being combined with many novel agents including anti-CTLA-4 antibodies, PARP inhibitors, targeted agents, and traditional chemotherapy in promising studies with the hopes of increasing the immune response and overcoming resistance by targeting other pathways. Novel immune techniques including vaccines and adoptive cell therapies are also being implemented in gynecological cancers. Immune checkpoint combinations and novel immunotherapy strategies have demonstrated potential to overcome resistance to PD-1/PD-L1 blockade in gynecological cancers. Identification of biomarkers of response and resistance is a priority to tailor specific combination therapies to the appropriate patients.
Collapse
Affiliation(s)
- Ying L Liu
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Dmitriy Zamarin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Weill Cornell Medical College, New York, NY, USA.
- Gynecologic Medical Oncology, Immunotherapeutics Service, Memorial Sloan-Kettering Cancer Center, 300 East 66th street, 1313, New York, NY, 10065, USA.
| |
Collapse
|
3241
|
Mou H, Yu L, Liao Q, Hou X, Wu Y, Cui Q, Yan N, Ma R, Wang L, Yao M, Wang K. Successful response to the combination of immunotherapy and chemotherapy in cholangiocarcinoma with high tumour mutational burden and PD-L1 expression: a case report. BMC Cancer 2018; 18:1105. [PMID: 30419854 PMCID: PMC6233589 DOI: 10.1186/s12885-018-5021-2] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 10/31/2018] [Indexed: 12/21/2022] Open
Abstract
Background Cholangiocarcinoma, or bile duct cancer, is a gastrointestinal cancer with limited therapeutic options and a poor outcome. Studies have revealed that some major driver genes are associated with cholangiocarcinoma, but no targeted therapies have been approved. Immune checkpoint inhibitors, which are represented by inhibitors of programmed cell death 1 (PD-1)/programmed death-ligand 1 (PD-L1), have emerged as a potential therapy for multiple types of solid cancers. Case presentation A 53-year-old female presented with postoperative recurrence of PD-L1-positive intrahepatic cholangiocarcinoma with a high tumour mutational burden. This patient exhibited a marked response to the combination of anti-PD-1 immunotherapy and chemotherapy. Conclusions As far as we know, this is the first case report on the success of the combination of immunotherapy and chemotherapy for advanced cholangiocarcinoma with PD-L1 positivity and a high tumour mutational burden.
Collapse
Affiliation(s)
- Haibo Mou
- Department of Medical Oncology, Shulan (Hangzhou) Hospital, #848 Dongxin Road, Hangzhou, Zhejiang Province, 310022, People's Republic of China.
| | - Lanfang Yu
- Department of Medical Oncology, Shulan (Hangzhou) Hospital, #848 Dongxin Road, Hangzhou, Zhejiang Province, 310022, People's Republic of China
| | - Qin Liao
- Department of Medical Oncology, Shulan (Hangzhou) Hospital, #848 Dongxin Road, Hangzhou, Zhejiang Province, 310022, People's Republic of China
| | - Xuehua Hou
- Department of Medical Oncology, Shulan (Hangzhou) Hospital, #848 Dongxin Road, Hangzhou, Zhejiang Province, 310022, People's Republic of China
| | - Yinfang Wu
- Department of Medical Oncology, Shulan (Hangzhou) Hospital, #848 Dongxin Road, Hangzhou, Zhejiang Province, 310022, People's Republic of China
| | - Qiang Cui
- OrigiMed, 115 Xinjun Huan Road, Minhang District, Shanghai, 201114, China
| | - Na Yan
- OrigiMed, 115 Xinjun Huan Road, Minhang District, Shanghai, 201114, China
| | - Ruobing Ma
- OrigiMed, 115 Xinjun Huan Road, Minhang District, Shanghai, 201114, China
| | - Lingjian Wang
- OrigiMed, 115 Xinjun Huan Road, Minhang District, Shanghai, 201114, China
| | - Ming Yao
- OrigiMed, 115 Xinjun Huan Road, Minhang District, Shanghai, 201114, China
| | - Kai Wang
- OrigiMed, 115 Xinjun Huan Road, Minhang District, Shanghai, 201114, China
| |
Collapse
|
3242
|
Hirsch L, Zitvogel L, Eggermont A, Marabelle A. PD-Loma: a cancer entity with a shared sensitivity to the PD-1/PD-L1 pathway blockade. Br J Cancer 2018; 120:3-5. [PMID: 30413824 PMCID: PMC6325162 DOI: 10.1038/s41416-018-0294-4] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 07/19/2018] [Accepted: 07/19/2018] [Indexed: 12/30/2022] Open
Abstract
Clinical trials have now identified over 30 cancer histotypes with sensitivity to anti-PD-(L)1 therapies. It is the first time in oncology that a class of drugs has demonstrated such a wide spectrum of activity in monotherapy. This subgroup of cancers (‘PD-Lomas’) is driving the clinical research strategies for the next generation of combination immunotherapy.
Collapse
Affiliation(s)
- Laure Hirsch
- Département d'Innovation Thérapeutique et d'Essais Précoces, Gustave Roussy, Université Paris-Saclay, Villejuif, F-94805, France
| | - Laurence Zitvogel
- Gustave Roussy, Université Paris-Saclay, INSERM U1015, Villejuif, F-94805, France
| | | | - Aurelien Marabelle
- Département d'Innovation Thérapeutique et d'Essais Précoces, Gustave Roussy, Université Paris-Saclay, Villejuif, F-94805, France. .,Gustave Roussy, Université Paris-Saclay, INSERM U1015, Villejuif, F-94805, France.
| |
Collapse
|
3243
|
Santoni M, Montironi R, Battelli N, Massari F. Reply to Michael Staehler, Dena Battle, Axel Bex, Hans Hammers, and Daniel George's Letter to the Editor re: Arnaud Méjean, Alain Ravaud, Simon Thezenas, et al. Sunitinib Alone or After Nephrectomy in Metastatic Renal-cell Carcinoma. Eur Urol 2018;74:842-3: Lymphocyte Phenotype and Timing of Radical Nephrectomy in Patients Treated with Immunocheckpoint Inhibitors for Renal Cell Carcinoma. Eur Urol 2018; 75:e64-e66. [PMID: 30391082 DOI: 10.1016/j.eururo.2018.10.031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 10/12/2018] [Indexed: 11/26/2022]
Affiliation(s)
| | - Rodolfo Montironi
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, Ancona, Italy
| | | | | |
Collapse
|
3244
|
Lopez-Beltran A, Henriques V, Cimadamore A, Santoni M, Cheng L, Gevaert T, Blanca A, Massari F, Scarpelli M, Montironi R. The Identification of Immunological Biomarkers in Kidney Cancers. Front Oncol 2018; 8:456. [PMID: 30450335 PMCID: PMC6225533 DOI: 10.3389/fonc.2018.00456] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 10/01/2018] [Indexed: 12/18/2022] Open
Abstract
The recent approval of several agents have revolutionized the scenario of therapeutic management of metastatic renal cell carcinoma (RCC) allowing us to reach important clinical end points with extended patients' survival. Actually, every new drug approved has represented an important step forward to the improvement of patient's survival. On the other hand, we now understand that RCC includes a large group of tumor entities, each of them with different genetic and mutational alterations, but also showing different clinical behavior; a reason behind the needs of subtype specific personalized approach to therapy of RCC. Immunotherapy is gradually becoming a key factor in the therapeutic algorithm for patients with locally advanced or metastatic RCC. Due to the combination of potent treatment success and potentially deadly adverse effects from immune checkpoint inhibitors (ICI), gathering prognostic and predictive information about FDA-indicated tumors seems to be prudent. Robust and reliable biomarkers are crucial for patient's selection of treatments with immunomodulatory drugs. PD-L1 expression is a poor prognostic factor and predictive of better responses from both PD-1 and PD-L1 inhibitors in a variety of tumor types including RCC. Each FDA approved PD-1/PD-L1 drug is paired with a PD-L1 Immunohistochemistry (IHC) assay. Thus, there is need for improved knowledge and application of PD-1/PD-L1 IHC biomarkers in daily practice. IHC staining appears in membranous fashion. The atezolizumab approved IHC assay is unique in that only immune cell staining is quantified for the use of this assay in RCC. A single biomarker for patient selection may not be feasible, given that immune responses are dynamic and evolve over time. Biomarker development for ICI drugs will likely require integration of multiple biologic components like PD-L1 expression, TILs and mutational load. New methodological approaches based on digital pathology may be relevant since they will allow recognition of the biomarker and to objectively quantitate its expression, and therefore might produce objective and reproducible cut-off assessment. Multidisciplinary approach is very much needed to fully develop the current and future value of ICI in clinical practice.
Collapse
Affiliation(s)
- Antonio Lopez-Beltran
- Department of Pathology and Surgery, Faculty of Medicine, Cordoba University, Cordoba, Spain
| | | | - Alessia Cimadamore
- Section of Pathological Anatomy, United Hospital, School of Medicine, Polytechnic University of the Marche Region, Ancona, Italy
| | | | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Thomas Gevaert
- Laboratory of Experimental Urology, Organ Systems, KU Leuven, Leuven, Belgium.,Department of Pathology, AZ Klina, Brasschaat, Belgium
| | - Ana Blanca
- Instituto Maimonides de Investigación Biomédica de Córdoba, Córdoba, Spain
| | | | - Marina Scarpelli
- Section of Pathological Anatomy, United Hospital, School of Medicine, Polytechnic University of the Marche Region, Ancona, Italy
| | - Rodolfo Montironi
- Section of Pathological Anatomy, United Hospital, School of Medicine, Polytechnic University of the Marche Region, Ancona, Italy
| |
Collapse
|
3245
|
Nuti M, Zizzari I, Botticelli A, Rughetti A, Marchetti P. The ambitious role of anti angiogenesis molecules: Turning a cold tumor into a hot one. Cancer Treat Rev 2018; 70:41-46. [DOI: 10.1016/j.ctrv.2018.07.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 07/23/2018] [Accepted: 07/25/2018] [Indexed: 12/29/2022]
|
3246
|
Dillman RO, Depriest C. Dendritic Cell Vaccines Presenting Autologous Tumor Antigens from Self-renewing Cancer Cells in Metastatic Renal Cell Carcinoma. JOURNAL OF EXPLORATORY RESEARCH IN PHARMACOLOGY 2018; 3:93-101. [DOI: 10.14218/jerp.2018.00012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
3247
|
Bensalah K, Albiges L, Bernhard JC, Bigot P, Bodin T, Boissier R, Correas JM, Gimel P, Hetet JF, Long JA, Nouhaud FX, Ouzaïd I, Rioux-Leclercq N, Méjean A. Recommandations françaises du Comité de Cancérologie de l’AFU – Actualisation 2018–2020 : prise en charge du cancer du rein. Prog Urol 2018; 28 Suppl 1:R5-R33. [DOI: 10.1016/j.purol.2019.01.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 09/15/2018] [Indexed: 12/15/2022]
|
3248
|
Iacovelli R, Ciccarese C, Bria E, Bimbatti D, Fantinel E, Mosillo C, Bisogno I, Brunelli M, Tortora G, Porta C. Immunotherapy versus standard of care in metastatic renal cell carcinoma. A systematic review and meta-analysis. Cancer Treat Rev 2018; 70:112-117. [DOI: 10.1016/j.ctrv.2018.08.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 08/10/2018] [Accepted: 08/17/2018] [Indexed: 12/18/2022]
|
3249
|
Signoretti S, Flaifel A, Chen YB, Reuter VE. Renal Cell Carcinoma in the Era of Precision Medicine: From Molecular Pathology to Tissue-Based Biomarkers. J Clin Oncol 2018; 36:JCO2018792259. [PMID: 30372384 PMCID: PMC6299340 DOI: 10.1200/jco.2018.79.2259] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Renal cell carcinoma (RCC) is not a single entity but includes various tumor subtypes that have been identified on the basis of either characteristic pathologic features or distinctive molecular changes. Clear cell RCC is the most common type of RCC and is characterized by dysregulation of the von Hippel Lindau/hypoxia-inducible factor pathway. Non-clear cell RCC represents a more heterogeneous group of tumors with diverse histopathologic and molecular features. In the past two decades, the improved understanding of the molecular landscape of RCC has led to the development of more effective therapies for metastatic RCC, which include both targeted agents and immune checkpoint inhibitors. Because only subsets of patients with metastatic RCC respond to a given treatment, predictive biomarkers are needed to guide treatment selection and sequence. In this review, we describe the key histologic features and molecular alterations of RCC subtypes and discuss emerging tissue-based biomarkers of response to currently available therapies for metastatic disease.
Collapse
Affiliation(s)
- Sabina Signoretti
- Sabina Signoretti and Abdallah Flaifel, Brigham and Women’s Hospital; Sabina Signoretti, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Ying-Bei Chen and Victor E. Reuter, Memorial Sloan Kettering Cancer Center; and Victor E. Reuter, Weill Cornell Medical College, New York, NY
| | - Abdallah Flaifel
- Sabina Signoretti and Abdallah Flaifel, Brigham and Women’s Hospital; Sabina Signoretti, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Ying-Bei Chen and Victor E. Reuter, Memorial Sloan Kettering Cancer Center; and Victor E. Reuter, Weill Cornell Medical College, New York, NY
| | - Ying-Bei Chen
- Sabina Signoretti and Abdallah Flaifel, Brigham and Women’s Hospital; Sabina Signoretti, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Ying-Bei Chen and Victor E. Reuter, Memorial Sloan Kettering Cancer Center; and Victor E. Reuter, Weill Cornell Medical College, New York, NY
| | - Victor E. Reuter
- Sabina Signoretti and Abdallah Flaifel, Brigham and Women’s Hospital; Sabina Signoretti, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Ying-Bei Chen and Victor E. Reuter, Memorial Sloan Kettering Cancer Center; and Victor E. Reuter, Weill Cornell Medical College, New York, NY
| |
Collapse
|
3250
|
Graham J, Dudani S, Heng DYC. Prognostication in Kidney Cancer: Recent Advances and Future Directions. J Clin Oncol 2018; 36:JCO2018790147. [PMID: 30372388 DOI: 10.1200/jco.2018.79.0147] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The most common type of cancer originating in the kidney is renal cell carcinoma (RCC). In both localized and advanced RCC, a number of clinical, pathologic, and molecular factors have been identified as having prognostic significance. In localized disease, risk stratification has traditionally involved the anatomic extent of disease, and several integrated scoring systems have been developed to help predict outcomes after definitive local therapy. In metastatic RCC, integrated prognostic models have also been established. These are used to stratify patients in contemporary clinical trials and to guide risk-directed treatment selection in clinical practice. Although many prognostic factors are common to both localized and advanced disease, there are some important distinctions. In both of these types of disease, the prognostic role of specific molecular and genomic alterations is an area of active investigation. In this review, we highlight the current staging systems and prognostic factors in localized and metastatic RCC. We also explore future directions in this area, including the expanding role of molecular biomarkers and their integration into the traditional prognostic models.
Collapse
Affiliation(s)
- Jeffrey Graham
- Jeffrey Graham and Daniel Y.C. Heng, University of Calgary, Calgary, Alberta; and Shaan Dudani, University of Ottawa, Ottawa, Ontario, Canada
| | - Shaan Dudani
- Jeffrey Graham and Daniel Y.C. Heng, University of Calgary, Calgary, Alberta; and Shaan Dudani, University of Ottawa, Ottawa, Ontario, Canada
| | - Daniel Y C Heng
- Jeffrey Graham and Daniel Y.C. Heng, University of Calgary, Calgary, Alberta; and Shaan Dudani, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|