151
|
Abstract
Immune modulatory treatment regimens, led by immune checkpoint inhibitors, have transformed the treatment of clear-cell renal cell carcinoma. First-in-class, the PD-1 inhibitor nivolumab improved overall survival in advanced renal cell carcinoma following prior anti-angiogenic therapy, an important shift in the management of clear-cell renal cell carcinoma. Further improvements of long-term outcomes will be driven by combinations in the first-line setting, including PD-1/PD-L1 associated with antiangiogenic therapies, or PD1/PD-L1 inhibitors with other immune checkpoint inhibitors such as anti-CTLA-4, anti-LAG-3 or TIM-3 targeted therapies. The first two randomized Phase 3 trials assessing these combinations have now challenged sunitinib in first-line setting. First, the CheckMate 214 trial demonstrated an objective response rate and overall survival benefit for the combination of nivolumab plus ipilimumab in the intermediate- and poor-risk patients. Second, the IMMotion 151 study demonstrated a progression-free survival benefit for the atezolizumab plus bevacizumab combination by investigator assessment. Further Phase 3 trials are awaited with tyrosine kinase and immune checkpoint inhibitor combinations. Clinical trials of immune checkpoint inhibitors are also actively investigated in the localized adjuvant or neoadjuvant setting. Nevertheless, the search for biomarkers along with new clinical trial designs will be crucial to better select the patients that may derive the greatest benefit from these advances. The continuing improvement of antitumor immunity comprehension and the emergence of new immune modulatory treatments will deeply change the management of renal cell carcinoma for the years to come.
Collapse
|
152
|
Wang Y, Zhou S, Yang F, Qi X, Wang X, Guan X, Shen C, Duma N, Vera Aguilera J, Chintakuntlawar A, Price KA, Molina JR, Pagliaro LC, Halfdanarson TR, Grothey A, Markovic SN, Nowakowski GS, Ansell SM, Wang ML. Treatment-Related Adverse Events of PD-1 and PD-L1 Inhibitors in Clinical Trials: A Systematic Review and Meta-analysis. JAMA Oncol 2019; 5:1008-1019. [PMID: 31021376 PMCID: PMC6487913 DOI: 10.1001/jamaoncol.2019.0393] [Citation(s) in RCA: 509] [Impact Index Per Article: 101.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
IMPORTANCE Programmed cell death (PD-1) and programmed cell death ligand 1 (PD-L1) inhibitors have been increasingly used in cancer therapy. Understanding the treatment-related adverse events of these drugs is critical for clinical practice. OBJECTIVE To evaluate the incidences of treatment-related adverse events of PD-1 and PD-L1 inhibitors and the differences between different drugs and cancer types. DATA SOURCES PubMed, Web of Science, Embase, and Scopus were searched from October 1, 2017, through December 15, 2018. STUDY SELECTION Published clinical trials on single-agent PD-1 and PD-L1 inhibitors with tabulated data on treatment-related adverse events were included. DATA EXTRACTION AND SYNTHESIS Trial name, phase, cancer type, PD-1 and PD-L1 inhibitor used, dose escalation, dosing schedule, number of patients, number of all adverse events, and criteria for adverse event reporting data were extracted from each included study, and bayesian multilevel regression models were applied for data analysis. MAIN OUTCOMES AND MEASURES Incidences of treatment-related adverse events and differences between different drugs and cancer types. RESULTS This systematic review and meta-analysis included 125 clinical trials involving 20 128 patients; 12 277 (66.0%) of 18 610 patients from 106 studies developed at least 1 adverse event of any grade (severity), and 2627 (14.0%) of 18 715 patients from 110 studies developed at least 1 adverse event of grade 3 or higher severity. The most common all-grade adverse events were fatigue (18.26%; 95% CI, 16.49%-20.11%), pruritus (10.61%; 95% CI, 9.46%-11.83%), and diarrhea (9.47%; 95% CI, 8.43%-10.58%). The most common grade 3 or higher adverse events were fatigue (0.89%; 95% CI, 0.69%-1.14%), anemia (0.78%; 95% CI, 0.59%-1.02%), and aspartate aminotransferase increase (0.75%; 95% CI, 0.56%-0.99%). Hypothyroidism (6.07%; 95% CI, 5.35%-6.85%) and hyperthyroidism (2.82%; 95% CI, 2.40%-3.29%) were the most frequent all-grade endocrine immune-related adverse events. Nivolumab was associated with higher mean incidences of all-grade adverse events compared with pembrolizumab (odds ratio [OR], 1.28; 95% CI, 0.97-1.79) and grade 3 or higher adverse events (OR, 1.30; 95% CI, 0.89-2.00). PD-1 inhibitors were associated with a higher mean incidence of grade 3 or higher adverse events compared with PD-L1 inhibitors (OR, 1.58; 95% CI, 1.00-2.54). CONCLUSIONS AND RELEVANCE Different PD-1 and PD-L1 inhibitors appear to have varying treatment-related adverse events; a comprehensive summary of the incidences of treatment-related adverse events in clinical trials provides an important guide for clinicians.
Collapse
Affiliation(s)
- Yucai Wang
- Division of Hematology, Mayo Clinic, Rochester, Minnesota
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota
| | - Shouhao Zhou
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston
- Department of Public Health Sciences, Pennsylvania State College of Medicine, Hershey
| | - Fang Yang
- Medical School of Nanjing University, Nanjing, China
- The Comprehensive Cancer Centre of Drum Tower Hospital, Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Xinyue Qi
- Department of Health Services Research, The University of Texas MD Anderson Cancer Center, Houston
| | - Xin Wang
- Medical School of Nanjing University, Nanjing, China
| | - Xiaoxiang Guan
- Medical School of Nanjing University, Nanjing, China
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chan Shen
- Department of Health Services Research, The University of Texas MD Anderson Cancer Center, Houston
| | - Narjust Duma
- Division of Hematology, Mayo Clinic, Rochester, Minnesota
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota
| | - Jesus Vera Aguilera
- Division of Hematology, Mayo Clinic, Rochester, Minnesota
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota
| | | | | | | | | | | | - Axel Grothey
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota
- West Cancer Center, The University of Tennessee, Memphis
| | | | | | | | - Michael L. Wang
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston
| |
Collapse
|
153
|
Suzuki S, Toyoma S, Tomizawa H, Yamada T, Iikawa N, Shiina K, Saito H, Koizumi K, Kawasaki Y, Yamada T. Efficacy of chemotherapy after progression with nivolumab in squamous cell carcinoma of the head and neck. Auris Nasus Larynx 2019; 47:485-488. [PMID: 31248647 DOI: 10.1016/j.anl.2019.06.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Revised: 05/31/2019] [Accepted: 06/12/2019] [Indexed: 10/26/2022]
Abstract
Nivolumab, a programmed death-1 (PD-1) inhibitor, has shown promising results against squamous cell carcinoma of the head and neck (SCCHN) in cases of recurrence or in a metastatic setting after platinum-based therapy. However, treatment alternatives for patients with nivolumab-refractory are limited, and a constant opinion is not provided. Recently, accumulating studies have demonstrated that chemotherapy after immune checkpoint inhibitor treatment may induce better objective responses in patients with advanced non-small cell lung cancer. However, there are few reports on the increased effect of chemotherapy after nivolumab treatment in SCCHN. Therefore, cases must be accumulated to identify patients with nivolumab-refractory SCCHN who may benefit from chemotherapy. Here, we present patients with SCCHN who exhibited a significant response to chemotherapy after nivolumab treatment.
Collapse
Affiliation(s)
- Shinsuke Suzuki
- Department of Otorhinolaryngology & Head and Neck Surgery, Akita University Graduate School of Medicine, Akita 010-8543, Japan.
| | - Satoshi Toyoma
- Department of Otorhinolaryngology & Head and Neck Surgery, Akita University Graduate School of Medicine, Akita 010-8543, Japan
| | - Hiroki Tomizawa
- Department of Otorhinolaryngology & Head and Neck Surgery, Akita University Graduate School of Medicine, Akita 010-8543, Japan
| | - Toshiki Yamada
- Department of Otorhinolaryngology & Head and Neck Surgery, Akita University Graduate School of Medicine, Akita 010-8543, Japan
| | - Nobuko Iikawa
- Department of Otorhinolaryngology & Head and Neck Surgery, Akita University Graduate School of Medicine, Akita 010-8543, Japan
| | - Kazuhiro Shiina
- Department of Otorhinolaryngology & Head and Neck Surgery, Akita University Graduate School of Medicine, Akita 010-8543, Japan
| | - Hidekazu Saito
- Department of Otorhinolaryngology & Head and Neck Surgery, Akita University Graduate School of Medicine, Akita 010-8543, Japan
| | - Koh Koizumi
- Department of Otorhinolaryngology & Head and Neck Surgery, Akita University Graduate School of Medicine, Akita 010-8543, Japan
| | - Yohei Kawasaki
- Department of Otorhinolaryngology & Head and Neck Surgery, Akita University Graduate School of Medicine, Akita 010-8543, Japan
| | - Takechiyo Yamada
- Department of Otorhinolaryngology & Head and Neck Surgery, Akita University Graduate School of Medicine, Akita 010-8543, Japan
| |
Collapse
|
154
|
Role of Biomarkers in Prediction of Response to Therapeutics in Metastatic Renal-Cell Carcinoma. Clin Genitourin Cancer 2019; 17:e454-e460. [DOI: 10.1016/j.clgc.2019.01.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 12/10/2018] [Accepted: 01/08/2019] [Indexed: 01/06/2023]
|
155
|
Differential Kinase Activation in Peripheral Blood Mononuclear Cells from Non-Small-Cell Lung Cancer Patients Treated with Nivolumab. Cancers (Basel) 2019; 11:cancers11060762. [PMID: 31159331 PMCID: PMC6628172 DOI: 10.3390/cancers11060762] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 05/22/2019] [Accepted: 05/25/2019] [Indexed: 12/24/2022] Open
Abstract
In the era of precision medicine, research of biomarkers for identification of responders to nivolumab therapy is a major challenge. Peripheral blood mononuclear cells (PBMC) could be an interesting surrogate tissue for identifying pharmacodynamic biomarkers. The aim of this exploratory study was to investigate the global serine/threonine kinase (STK) activity in PBMC from non-small-cell lung cancer (NSCLC) patients using a high throughput kinomic profiling method. PamChip® microarrays were used to explore the STK kinomic profile in PBMC from 28 NSCLC patients before nivolumab initiation (D0) and on day 14 (D14) of the first administration. Two clusters of patients (A and B) were identified at D0, median overall survival (OS) tended to be longer in cluster A than in B (402 vs. 112.5 days, respectively; p = 0.15). Interestingly, the PD-L1 tumor cell score (p = 0.045), the count of CD8+ cells (p = 0.023) and the total body weight (p = 0.038) were statistically different between the clusters. On D14, clusters C and D were identified. Greater activity of most STK, especially those of the PI3K/Akt signaling pathway, was noticed among cluster C. No significant difference between C and D was observed regarding OS. Considering the small number of patients, results from this preliminary study are not conclusive. However, the 4-fold longer median OS in cluster A paves the way to further investigate, in a larger cohort of NSCLC patients, the benefit of basal STK kinomic profile in PBMC to identify responders to nivolumab therapy.
Collapse
|
156
|
Seliger B. Combinatorial Approaches With Checkpoint Inhibitors to Enhance Anti-tumor Immunity. Front Immunol 2019; 10:999. [PMID: 31178856 PMCID: PMC6538766 DOI: 10.3389/fimmu.2019.00999] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 04/18/2019] [Indexed: 12/13/2022] Open
Abstract
Treatment of cancer patients has been recently revolutionized by the application of various immunotherapeutics. However, the response rates are still limited ranging between approximately 20 and 40% suggesting that combinations of immunotherapy with conventional treatment, like chemotherapy, radiation, epigenetic modulators, targeted therapies using small molecules as well as other (immuno) therapeutics, might be an option to increase systemic anti-tumor immunity. It is postulated that different non-immune based therapies in combination with immunotherapies could reprogram the immune suppressive tumor microenvironment and enhance the immunogenicity of tumor cells leading to an improved therapeutic efficacy and a better patients' outcome. Despite there exist various examples of increased objective responses achieved by adding these different therapies to immunotherapies, strategies for rational and evidence-based design of checkpoint inhibitor combinations to maximize the clinical benefit for patients are urgently required. Therefore, the main purpose of this review is to summarize recent results obtained from experimental models and clinical trials to enhance tumor immunogenicity by combining immunotherapy with other therapeutic options to maximize patients' outcome and minimize adverse events.
Collapse
Affiliation(s)
- Barbara Seliger
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| |
Collapse
|
157
|
Saleh R, Elkord E. Treg-mediated acquired resistance to immune checkpoint inhibitors. Cancer Lett 2019; 457:168-179. [PMID: 31078738 DOI: 10.1016/j.canlet.2019.05.003] [Citation(s) in RCA: 146] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 04/29/2019] [Accepted: 05/06/2019] [Indexed: 02/08/2023]
Abstract
T Regulatory cells (Tregs) act as a double-edged sword by regulating immune homeostasis (protective role) and inhibiting immune responses in different disease settings (pathological role). They contribute to cancer development and progression by suppressing T effector cell (Teff) functions. Decreased ratios of intratumoral CD8+ T cells to Tregs have been associated with poor prognosis in most cancer types. Targeting immune checkpoints (ICs), such as cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) and programmed cell death-1 (PD-1), by immune checkpoint inhibitors (ICIs) in cancer patients has been beneficial in inducing anti-tumor immune responses and improving clinical outcomes. However, response rates remain relatively low, ranging from 15 to 40% depending on cancer type. Additionally, a significant proportion of patients who initially demonstrates a clinical response can acquire resistance overtime. This acquired resistance could occur due to the emergence of compensatory mechanisms within the tumor microenvironment (TME) to evade the anti-tumor effects of ICIs. In this review, we describe the immunosuppressive role of Tregs in the TME, the effects of currently approved ICIs on Treg phenotype and function, and the mechanisms of acquired resistance to ICIs mediated by Tregs within the TME, such as the over-expression of ICs, the up-regulation of immunosuppressive molecules, and apoptotic Treg-induced immunosuppression. We also describe potential therapeutic strategies to target Tregs in combination with ICIs aiming to overcome such resistance and improve clinical outcomes. Elucidating the Treg-mediated acquired resistance mechanisms should benefit the designing of well-targeted therapeutic strategies to overcome resistance and maximize the therapeutic efficacy in cancer patients.
Collapse
Affiliation(s)
- Reem Saleh
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Eyad Elkord
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar.
| |
Collapse
|
158
|
Shah AY, Kotecha RR, Lemke EA, Chandramohan A, Chaim JL, Msaouel P, Xiao L, Gao J, Campbell MT, Zurita AJ, Wang J, Corn PG, Jonasch E, Motzer RJ, Sharma P, Voss MH, Tannir NM. Outcomes of patients with metastatic clear-cell renal cell carcinoma treated with second-line VEGFR-TKI after first-line immune checkpoint inhibitors. Eur J Cancer 2019; 114:67-75. [PMID: 31075726 DOI: 10.1016/j.ejca.2019.04.003] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 03/29/2019] [Accepted: 04/02/2019] [Indexed: 01/05/2023]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) are being increasingly utilised in the front-line (1L) setting of metastatic clear-cell renal cell carcinoma (mccRCC). Limited data exist on responses and survival on second-line (2L) vascular endothelial growth factor-receptor tyrosine kinase inhibitor (VEGFR-TKI) therapy after 1L ICI therapy. PATIENTS AND METHODS This is a retrospective study of mccRCC patients treated with 2L VEGFR-TKI after progressive disease (PD) with 1L ICI. Patients were treated at MD Anderson Cancer Center or Memorial Sloan Kettering Cancer Center between December 2015 and February 2018. Objective response was assessed by blinded radiologists' review using Response Evaluation Criteria in Solid Tumours v1.1. Descriptive statistics and Kaplan-Meier method were used. RESULTS Seventy patients were included in the analysis. Median age at mccRCC diagnosis was 59 years; 8 patients (11%) had international metastatic database consortium favourable-risk disease, 48 (69%) had intermediate-risk disease and 14 (20%) had poor-risk disease. As 1L therapy, 12 patients (17%) received anti-programmed death ligand-1 (PD-(L)1) monotherapy with nivolumab or atezolizumab, 33 (47%) received nivolumab plus ipilimumab and 25 (36%) received combination anti-PD-(L)1 plus bevacizumab. 2L TKI therapies included pazopanib, sunitinib, axitinib and cabozantinib. On 2L TKI therapy, one patient (1.5%) achieved a complete response, 27 patients (39.7%) a partial response and 36 patients (52.9%) stable disease. Median progression-free survival (mPFS) was 13.2 months (95% confidence interval: 10.1, NA). Forty-five percent of subjects required a dose reduction, and twenty-seven percent of patients discontinued treatment because of toxicity. CONCLUSIONS In this retrospective study of patients with mccRCC receiving 2L TKI monotherapy after 1L ICI, we observed 2L antitumour activity and tolerance comparable to historical data for 1L TKI.
Collapse
Affiliation(s)
- A Y Shah
- Department of Genitourinary Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA.
| | - R R Kotecha
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - E A Lemke
- Department of Genitourinary Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - A Chandramohan
- Department of Diagnostic Radiology, MD Anderson Cancer Center, Houston, TX, USA
| | - J L Chaim
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - P Msaouel
- Department of Genitourinary Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - L Xiao
- Department of Biostatistics, MD Anderson Cancer Center, Houston, TX, USA
| | - J Gao
- Department of Genitourinary Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - M T Campbell
- Department of Genitourinary Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - A J Zurita
- Department of Genitourinary Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - J Wang
- Department of Genitourinary Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - P G Corn
- Department of Genitourinary Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - E Jonasch
- Department of Genitourinary Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - R J Motzer
- Genitourinary Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - P Sharma
- Department of Genitourinary Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - M H Voss
- Genitourinary Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - N M Tannir
- Department of Genitourinary Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
159
|
Cantara S, Bertelli E, Occhini R, Regoli M, Brilli L, Pacini F, Castagna MG, Toti P. Blockade of the programmed death ligand 1 (PD-L1) as potential therapy for anaplastic thyroid cancer. Endocrine 2019; 64:122-129. [PMID: 30762153 DOI: 10.1007/s12020-019-01865-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 02/05/2019] [Indexed: 02/06/2023]
Abstract
PURPOSE Anaplastic thyroid carcinoma (ATC) is a rare, highly aggressive form of thyroid cancer (TC) characterized by an aggressive behavior and poor prognosis, resulting in patients' death within a year. Standard treatments, such as chemo and radiotherapy, as well as tyrosine kinase inhibitors, are ineffective for ATC treatment. Cancer immunotherapy is one of the most promising research area in oncology. The PD-1/PD-L1 axis is of particular interest, in light of promising data showing a restoration of host immunity against tumors, with the prospect of long-lasting remissions. METHODS In this study, we evaluated PD-L1 expression in a large series of TCs (20 cases) showing a progressive dedifferentiation of the thyroid tumor from well differentiated TC to ATC, employing two different antibodies [R&D Systems and VENTANA PD-L1 (SP263) Rabbit Monoclonal Primary Antibody]. We also tested the anti PD-L1 mAb in an in vivo animal model. RESULTS We found that approximately 70-90% of ATC cases were positive for PD-L1 whereas normal thyroid and differentiated TC were negative. Moreover, all analyzed cases presented immunopositive staining in the endothelium of vessels within or in close proximity to the tumor, while normal thyroid vessels were negative. PD-L1 mAb was also effective in inhibiting ATC growth in an in vivo model. CONCLUSIONS These data suggest that immunotherapy may be a promising treatment specific for ATC suggesting the need to start with clinical TRIALs.
Collapse
Affiliation(s)
- Silvia Cantara
- Department of Medical, Surgical and Neurological Sciences, University of Siena, Siena, Italy.
| | - Eugenio Bertelli
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | | | - Marì Regoli
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Lucia Brilli
- Department of Medical, Surgical and Neurological Sciences, University of Siena, Siena, Italy
| | - Furio Pacini
- Department of Medical, Surgical and Neurological Sciences, University of Siena, Siena, Italy
| | - Maria Grazia Castagna
- Department of Medical, Surgical and Neurological Sciences, University of Siena, Siena, Italy
| | | |
Collapse
|
160
|
Lenvatinib plus anti-PD-1 antibody combination treatment activates CD8+ T cells through reduction of tumor-associated macrophage and activation of the interferon pathway. PLoS One 2019. [PMID: 30811474 DOI: 10.1371/journal.pone.0212513.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Lenvatinib is a multiple receptor tyrosine kinase inhibitor targeting mainly vascular endothelial growth factor (VEGF) and fibroblast growth factor (FGF) receptors. We investigated the immunomodulatory activities of lenvatinib in the tumor microenvironment and its mechanisms of enhanced antitumor activity when combined with a programmed cell death-1 (PD-1) blockade. Antitumor activity was examined in immunodeficient and immunocompetent mouse tumor models. Single-cell analysis, flow cytometric analysis, and immunohistochemistry were used to analyze immune cell populations and their activation. Gene co-expression network analysis and pathway analysis using RNA sequencing data were used to identify lenvatinib-driven combined activity with anti-PD-1 antibody (anti-PD-1). Lenvatinib showed potent antitumor activity in the immunocompetent tumor microenvironment compared with the immunodeficient tumor microenvironment. Antitumor activity of lenvatinib plus anti-PD-1 was greater than that of either single treatment. Flow cytometric analysis revealed that lenvatinib reduced tumor-associated macrophages (TAMs) and increased the percentage of activated CD8+ T cells secreting interferon (IFN)-γ+ and granzyme B (GzmB). Combination treatment further increased the percentage of T cells, especially CD8+ T cells, among CD45+ cells and increased IFN-γ+ and GzmB+ CD8+ T cells. Transcriptome analyses of tumors resected from treated mice showed that genes specifically regulated by the combination were significantly enriched for type-I IFN signaling. Pretreatment with lenvatinib followed by anti-PD-1 treatment induced significant antitumor activity compared with anti-PD-1 treatment alone. Our findings show that lenvatinib modulates cancer immunity in the tumor microenvironment by reducing TAMs and, when combined with PD-1 blockade, shows enhanced antitumor activity via the IFN signaling pathway. These findings provide a scientific rationale for combination therapy of lenvatinib with PD-1 blockade to improve cancer immunotherapy.
Collapse
|
161
|
Kato Y, Tabata K, Kimura T, Yachie-Kinoshita A, Ozawa Y, Yamada K, Ito J, Tachino S, Hori Y, Matsuki M, Matsuoka Y, Ghosh S, Kitano H, Nomoto K, Matsui J, Funahashi Y. Lenvatinib plus anti-PD-1 antibody combination treatment activates CD8+ T cells through reduction of tumor-associated macrophage and activation of the interferon pathway. PLoS One 2019; 14:e0212513. [PMID: 30811474 PMCID: PMC6392299 DOI: 10.1371/journal.pone.0212513] [Citation(s) in RCA: 312] [Impact Index Per Article: 62.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 02/04/2019] [Indexed: 12/13/2022] Open
Abstract
Lenvatinib is a multiple receptor tyrosine kinase inhibitor targeting mainly vascular endothelial growth factor (VEGF) and fibroblast growth factor (FGF) receptors. We investigated the immunomodulatory activities of lenvatinib in the tumor microenvironment and its mechanisms of enhanced antitumor activity when combined with a programmed cell death-1 (PD-1) blockade. Antitumor activity was examined in immunodeficient and immunocompetent mouse tumor models. Single-cell analysis, flow cytometric analysis, and immunohistochemistry were used to analyze immune cell populations and their activation. Gene co-expression network analysis and pathway analysis using RNA sequencing data were used to identify lenvatinib-driven combined activity with anti-PD-1 antibody (anti-PD-1). Lenvatinib showed potent antitumor activity in the immunocompetent tumor microenvironment compared with the immunodeficient tumor microenvironment. Antitumor activity of lenvatinib plus anti-PD-1 was greater than that of either single treatment. Flow cytometric analysis revealed that lenvatinib reduced tumor-associated macrophages (TAMs) and increased the percentage of activated CD8+ T cells secreting interferon (IFN)-γ+ and granzyme B (GzmB). Combination treatment further increased the percentage of T cells, especially CD8+ T cells, among CD45+ cells and increased IFN-γ+ and GzmB+ CD8+ T cells. Transcriptome analyses of tumors resected from treated mice showed that genes specifically regulated by the combination were significantly enriched for type-I IFN signaling. Pretreatment with lenvatinib followed by anti-PD-1 treatment induced significant antitumor activity compared with anti-PD-1 treatment alone. Our findings show that lenvatinib modulates cancer immunity in the tumor microenvironment by reducing TAMs and, when combined with PD-1 blockade, shows enhanced antitumor activity via the IFN signaling pathway. These findings provide a scientific rationale for combination therapy of lenvatinib with PD-1 blockade to improve cancer immunotherapy.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antineoplastic Agents/administration & dosage
- CD8-Positive T-Lymphocytes/drug effects
- CD8-Positive T-Lymphocytes/immunology
- Cell Line, Tumor
- Gene Expression/drug effects
- Gene Expression/immunology
- Immunologic Factors/administration & dosage
- Interferons/metabolism
- Lymphocyte Activation/drug effects
- Macrophages/drug effects
- Macrophages/immunology
- Melanoma, Experimental/genetics
- Melanoma, Experimental/immunology
- Melanoma, Experimental/therapy
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Nude
- Neoplasms, Experimental/genetics
- Neoplasms, Experimental/immunology
- Neoplasms, Experimental/therapy
- Phenylurea Compounds/administration & dosage
- Programmed Cell Death 1 Receptor/antagonists & inhibitors
- Protein Kinase Inhibitors/administration & dosage
- Quinolines/administration & dosage
- Signal Transduction/drug effects
- Tumor Microenvironment/drug effects
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- Yu Kato
- Tsukuba Research Laboratories, Eisai Co., Ltd., Tsukuba, Ibaraki, Japan
| | - Kimiyo Tabata
- Tsukuba Research Laboratories, Eisai Co., Ltd., Tsukuba, Ibaraki, Japan
| | - Takayuki Kimura
- Tsukuba Research Laboratories, Eisai Co., Ltd., Tsukuba, Ibaraki, Japan
| | | | - Yoichi Ozawa
- Tsukuba Research Laboratories, Eisai Co., Ltd., Tsukuba, Ibaraki, Japan
| | - Kazuhiko Yamada
- Tsukuba Research Laboratories, Eisai Co., Ltd., Tsukuba, Ibaraki, Japan
| | - Junichi Ito
- Tsukuba Research Laboratories, Eisai Co., Ltd., Tsukuba, Ibaraki, Japan
| | - Sho Tachino
- Tsukuba Research Laboratories, Eisai Co., Ltd., Tsukuba, Ibaraki, Japan
| | - Yusaku Hori
- Tsukuba Research Laboratories, Eisai Co., Ltd., Tsukuba, Ibaraki, Japan
| | - Masahiro Matsuki
- Tsukuba Research Laboratories, Eisai Co., Ltd., Tsukuba, Ibaraki, Japan
| | | | - Samik Ghosh
- The Systems Biology Institute, Shinagawa, Tokyo, Japan
| | | | - Kenichi Nomoto
- Oncology Business Group, Eisai Inc., Woodcliff Lake, New Jersey, United States of America
| | - Junji Matsui
- Tsukuba Research Laboratories, Eisai Co., Ltd., Tsukuba, Ibaraki, Japan
- Oncology Business Group, Eisai Inc., Woodcliff Lake, New Jersey, United States of America
| | | |
Collapse
|
162
|
Denaro N, Merlano MC. Unexpected response with palliative conventional therapy in head and neck squamous cell carcinoma after anti-programmed death-1 progression. Head Neck 2019; 41:E42-E47. [PMID: 30614126 DOI: 10.1002/hed.25418] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 07/11/2018] [Accepted: 09/12/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The application of anti-programmed death-1 (PD-1) therapies for recurrent or metastatic head and neck squamous cell carcinoma (HNSCC) has found promising results. It is the first "second-line therapy" in this setting to impact on prognosis. The studies demonstrated overall response rates in the range of 20%. Therefore, HNSCC showed 60%-80% progression at first evaluation with better overall survival, suggesting regained efficacy of treatments given thereafter. METHODS We report three clinical cases treated with anti-PD-1 after platinum-based chemotherapy. RESULTS Our case reports achieved an unforeseen response to conventional therapies supporting the hypothesis of restored responses to conventional therapies after immunotherapy. CONCLUSIONS We believe that the inhibition of the PD-1/PD-ligand 1 checkpoint may synergize with both chemotherapy or radiotherapy through immunologic interplay, reversing the HNSCC-induced immune suppression.
Collapse
Affiliation(s)
- Nerina Denaro
- Oncology Department, ASO Santa Croce e Carle Cuneo, Cuneo, Italy
| | | |
Collapse
|
163
|
|
164
|
Yan C, Chang J, Song X, Yan F, Yu W, An Y, Wei F, Yang L, Ren X. Memory stem T cells generated by Wnt signaling from blood of human renal clear cell carcinoma patients. Cancer Biol Med 2019; 16:109-124. [PMID: 31119051 PMCID: PMC6528452 DOI: 10.20892/j.issn.2095-3941.2018.0118] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Objective Memory stem T cells (Tscm) have attracted attention because of their enhanced self-renewal, multipotent capacity, and anti-tumor capacities. However, little is known about Tscm in patients with renal clear cell carcinoma (RCC) and the role of Wnt signaling in these cells. We evaluated Tscm from RCC patients concerning their activation of Wnt signaling in vitro and explored the mechanism of preferential survival.
Methods Flow cytometry identified surface markers and cytokines produced from accumulated Tscm in the presence of the glycogen synthase kinase beta inhibitor TWS119. Apoptosis was evaluated after induction using tumor necrosis factor-alpha. Immunofluorescence and Western blot analyses were used to investigate the activation of the nuclear factor-kappa B (NF-КB) pathway. Results RCC patients had a similar percentage of CD4+ and CD8+ Tscm as healthy donors. Activation of Wnt signaling by TWS119 resulted in the accumulation of Tscm in activated T cells, but reversal of differentiated T cells to Tscm was not achieved. Preferential survival of Tscm was associated with increased anti-apoptotic ability mediated downstream of the NF-КB activation pathway.
Conclusions The finding that Tscm can accumulate by Wnt signaling in vitro in blood from RCC patients will help in devising new cancer therapy strategies of Tscm-based adoptive immunotherapy, such as dendritic cell-stimulated Tscm, and T cell receptor or chimeric antigen receptor-engineered Tscm.
Collapse
Affiliation(s)
- Cihui Yan
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Jingjing Chang
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Xinmiao Song
- Department of Electromyogram, 3rd Affiliated Hospital of Hebei Medical University, Shijiazhuang 050051, China
| | - Fan Yan
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Wenwen Yu
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Yang An
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Feng Wei
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Lili Yang
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Xiubao Ren
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| |
Collapse
|
165
|
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: 49] [Impact Index Per Article: 8.2] [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
|
166
|
Manoharan M, Mandloi N, Priyadarshini S, Patil A, Gupta R, Iyer L, Gupta R, Chaudhuri A. A Computational Approach Identifies Immunogenic Features of Prognosis in Human Cancers. Front Immunol 2018; 9:3017. [PMID: 30622534 PMCID: PMC6308325 DOI: 10.3389/fimmu.2018.03017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Accepted: 12/06/2018] [Indexed: 12/25/2022] Open
Abstract
A large number of tumor intrinsic and extrinsic factors determine long-term survival in human cancers. In this study, we stratified 9120 tumors from 33 cancers with respect to their immune cell content and identified immunogenomic features associated with long-term survival. Our analysis demonstrates that tumors infiltrated by CD8+ T cells expressing higher levels of activation marker (PD1hi) along with TCR signaling genes and cytolytic T cell markers (IL2hi/TNF-αhi/IFN-γhi/GZMA-Bhi) extend survival, whereas survival benefit was absent for tumors infiltrated by anergic and hyperexhausted CD8+ T cells characterized by high expression of CTLA-4, TIM3, LAG3, and genes linked to PI3K signaling pathway. The computational approach of using robust and highly specific gene expression signatures to deconvolute the tumor microenvironment has important clinical applications, such as selecting patients who will benefit from checkpoint inhibitor treatment.
Collapse
|
167
|
Pan M, Alavi M, Herrinton LJ. Association of Inflammatory Markers with Disease Progression in Patients with Metastatic Melanoma Treated with Immune Checkpoint Inhibitors. Perm J 2018; 22:17-149. [PMID: 29616914 DOI: 10.7812/tpp/17-149] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
INTRODUCTION We investigated the effect of inflammatory biomarkers (neutrophil, platelet, and lymphocyte counts) on risk of progression in patients with metastatic melanoma treated with an immune checkpoint inhibitor targeting programmed cell death protein-1 (PD-1). METHODS This retrospective cohort study included 108 patients with malignant melanoma treated with an anti-PD-1 checkpoint inhibitor from August 2014 through December 2015. The outcome was disease progression noted on imaging or clinical examination. Follow-up began on the date of initiation of anti-PD-1 therapy and ended on the date of progression, disenrollment, death of causes other than malignant melanoma, or the end of the study in February 2017. RESULTS The median time from initiating therapy with an anti-PD-1 checkpoint inhibitor (nivolumab or pembrolizumab) to the end of follow-up was 118 days. After adjustment, baseline neutrophil and platelet counts were associated with progression. The hazard ratio (HR) for neutrophil counts ≥ 5501/μL vs ≤ 3900/μL was 2.3 (95% confidence interval [CI] = 1.2-4.6, p < 0.05). For platelet counts ≥ 304,000 vs ≤ 215,000/μL, the HR was 2.0 (CI = 1.0-3.9, p < 0.05). For lymphocyte counts ≥ 1716/μL vs ≤ 1120/μL, the HR was 0.5 (CI = 0.2-1.0, p = 0.05). CONCLUSION For patients with metastatic melanoma treated with nivolumab or pembrolizumab, higher neutrophil or platelet counts, or lower lymphocyte counts, are associated with higher risk of progression. For these patients, we recommend more frequent assessment for progression and closer follow-up, especially for patients with substantial comorbidities or poor physical performance.
Collapse
Affiliation(s)
- Minggui Pan
- Medical Oncologist at the Santa Clara Medical Center and an Adjunct Investigator for the Division of Research in Oakland, CA.
| | | | | |
Collapse
|
168
|
Biomarqueurs prédictifs de réponse aux inhibiteurs de points de contrôle immuns. Bull Cancer 2018; 105 Suppl 1:S80-S91. [DOI: 10.1016/s0007-4551(18)30393-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 10/23/2017] [Indexed: 11/21/2022]
|
169
|
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: 0.5] [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
|
170
|
Shirotake S, Kaneko G, Nagata K, Oyama M, Nishimoto K. Histological complete response with nivolumab for renal cell carcinoma with multiple metastases: A case report. Mol Clin Oncol 2018; 10:244-248. [PMID: 30680202 DOI: 10.3892/mco.2018.1779] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 10/05/2018] [Indexed: 01/05/2023] Open
Abstract
The present case report describes a case of left renal clear cell carcinoma with brain, lung, para-aortic, and lymph node metastases (cT1bN1M1) in a 52-year-old Japanese male. The patient received sequential anticancer treatments with pazopanib, everolimus, and axitinib, but exhibited treatment-resistant tumor growth. Treatment with nivolumab resulted in a complete response in metastatic sites. However, the residual renal tumor, which was enhanced by contrast medium, required radical nephrectomy. Pathological analyses of the renal tumor revealed that it consisted of fibrotic and lymphocyte-infiltrated tissues in which morphological cancer cells were not detected. The majority of lymphocytes were cluster of differentiation (CD)8-positive, suggesting that cancer cells were attacked by these lymphocytes. Retrospective analyses of renal cell carcinoma tissues, which were biopsied before the anticancer treatment, revealed their infiltration by CD8-positive T cells. To the best of our knowledge, this is the first case report to examine renal tissue prior to and following treatment with nivolumab using immunohistochemical analysis.
Collapse
Affiliation(s)
- Suguru Shirotake
- Department of Uro-Oncology, Saitama Medical University International Medical Center, Saitama 350-1298, Japan
| | - Go Kaneko
- Department of Uro-Oncology, Saitama Medical University International Medical Center, Saitama 350-1298, Japan
| | - Koji Nagata
- Department of Diagnostic Pathology, Saitama Medical University International Medical Center, Saitama 350-1298, Japan
| | - Masafumi Oyama
- Department of Uro-Oncology, Saitama Medical University International Medical Center, Saitama 350-1298, Japan
| | - Koshiro Nishimoto
- Department of Uro-Oncology, Saitama Medical University International Medical Center, Saitama 350-1298, Japan
| |
Collapse
|
171
|
Wong YNS, Joshi K, Khetrapal P, Ismail M, Reading JL, Sunderland MW, Georgiou A, Furness AJS, Ben Aissa A, Ghorani E, Oakes T, Uddin I, Tan WS, Feber A, McGovern U, Swanton C, Freeman A, Marafioti T, Briggs TP, Kelly JD, Powles T, Peggs KS, Chain BM, Linch MD, Quezada SA. Urine-derived lymphocytes as a non-invasive measure of the bladder tumor immune microenvironment. J Exp Med 2018; 215:2748-2759. [PMID: 30257862 PMCID: PMC6219732 DOI: 10.1084/jem.20181003] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 08/03/2018] [Accepted: 09/05/2018] [Indexed: 12/19/2022] Open
Abstract
Despite the advances in cancer immunotherapy, only a fraction of patients with bladder cancer exhibit responses to checkpoint blockade, highlighting a need to better understand drug resistance and identify rational immunotherapy combinations. However, accessibility to the tumor prior and during therapy is a major limitation in understanding the immune tumor microenvironment (TME). Herein, we identified urine-derived lymphocytes (UDLs) as a readily accessible source of T cells in 32 patients with muscle invasive bladder cancer (MIBC). We observed that effector CD8+ and CD4+ cells and regulatory T cells within the urine accurately map the immune checkpoint landscape and T cell receptor repertoire of the TME. Finally, an increased UDL count, specifically high expression of PD-1 (PD-1hi) on CD8+ at the time of cystectomy, was associated with a shorter recurrence-free survival. UDL analysis represents a dynamic liquid biopsy that is representative of the bladder immune TME that may be used to identify actionable immuno-oncology (IO) targets with potential prognostic value in MIBC.
Collapse
Affiliation(s)
- Yien Ning Sophia Wong
- Cancer Immunology Unit, University College London (UCL) Cancer Institute, London, England, UK
- Research Department of Haematology, UCL Cancer Institute, London, England, UK
- Department of Oncology, UCL Cancer Institute, London, England, UK
- Department of Oncology, University College London Hospital, London, England, UK
| | - Kroopa Joshi
- Cancer Immunology Unit, University College London (UCL) Cancer Institute, London, England, UK
- Research Department of Haematology, UCL Cancer Institute, London, England, UK
- Division of Infection and Immunity, University College London, London, England, UK
- Department of Medical Oncology, The Royal Marsden NHS Foundation Trust, London, England, UK
| | - Pramit Khetrapal
- Department of Urology, University College London Hospital at Westmoreland Street, London, England, UK
- Division of Surgical and Interventional Sciences, University College London, London, England, UK
| | - Mazlina Ismail
- Division of Infection and Immunity, University College London, London, England, UK
| | - James L Reading
- Cancer Immunology Unit, University College London (UCL) Cancer Institute, London, England, UK
- Research Department of Haematology, UCL Cancer Institute, London, England, UK
| | - Mariana Werner Sunderland
- Cancer Immunology Unit, University College London (UCL) Cancer Institute, London, England, UK
- Research Department of Haematology, UCL Cancer Institute, London, England, UK
| | - Andrew Georgiou
- Cancer Immunology Unit, University College London (UCL) Cancer Institute, London, England, UK
- Research Department of Haematology, UCL Cancer Institute, London, England, UK
| | - Andrew J S Furness
- Cancer Immunology Unit, University College London (UCL) Cancer Institute, London, England, UK
- Research Department of Haematology, UCL Cancer Institute, London, England, UK
- Department of Medical Oncology, The Royal Marsden NHS Foundation Trust, London, England, UK
| | - Assma Ben Aissa
- Cancer Immunology Unit, University College London (UCL) Cancer Institute, London, England, UK
- Research Department of Haematology, UCL Cancer Institute, London, England, UK
| | - Ehsan Ghorani
- Cancer Immunology Unit, University College London (UCL) Cancer Institute, London, England, UK
- Research Department of Haematology, UCL Cancer Institute, London, England, UK
| | - Theres Oakes
- Division of Infection and Immunity, University College London, London, England, UK
| | - Imran Uddin
- Division of Infection and Immunity, University College London, London, England, UK
| | - Wei Shen Tan
- Department of Urology, University College London Hospital at Westmoreland Street, London, England, UK
- Division of Surgical and Interventional Sciences, University College London, London, England, UK
| | - Andrew Feber
- Division of Surgical and Interventional Sciences, University College London, London, England, UK
| | - Ursula McGovern
- Department of Oncology, University College London Hospital, London, England, UK
| | | | - Alex Freeman
- Department of Cellular Pathology, University College London Hospital, London, England, UK
| | - Teresa Marafioti
- Department of Cellular Pathology, University College London Hospital, London, England, UK
| | - Timothy P Briggs
- Department of Urology, University College London Hospital at Westmoreland Street, London, England, UK
| | - John D Kelly
- Department of Urology, University College London Hospital at Westmoreland Street, London, England, UK
- Division of Surgical and Interventional Sciences, University College London, London, England, UK
| | - Thomas Powles
- Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London, London, England, UK
| | - Karl S Peggs
- Cancer Immunology Unit, University College London (UCL) Cancer Institute, London, England, UK
- Research Department of Haematology, UCL Cancer Institute, London, England, UK
| | - Benjamin M Chain
- Division of Infection and Immunity, University College London, London, England, UK
| | - Mark D Linch
- Department of Oncology, UCL Cancer Institute, London, England, UK
- Department of Oncology, University College London Hospital, London, England, UK
| | - Sergio A Quezada
- Cancer Immunology Unit, University College London (UCL) Cancer Institute, London, England, UK
- Research Department of Haematology, UCL Cancer Institute, London, England, UK
| |
Collapse
|
172
|
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: 36] [Impact Index Per Article: 6.0] [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
|
173
|
McDermott DF, Carducci M. Progress in Kidney Cancer Outcomes Through Collaboration, Innovation, and Discovery. J Clin Oncol 2018; 36:JCO1801198. [PMID: 30372393 PMCID: PMC6299339 DOI: 10.1200/jco.18.01198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/06/2023] Open
Affiliation(s)
- David F. McDermott
- David F. McDermott, Dana-Farber/Harvard Cancer Center, Boston, MA; and Michael Carducci, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD
| | - Michael Carducci
- David F. McDermott, Dana-Farber/Harvard Cancer Center, Boston, MA; and Michael Carducci, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD
| |
Collapse
|
174
|
Amin A, Plimack ER, Ernstoff MS, Lewis LD, Bauer TM, McDermott DF, Carducci M, Kollmannsberger C, Rini BI, Heng DYC, Knox J, Voss MH, Spratlin J, Berghorn E, Yang L, Hammers HJ. Safety and efficacy of nivolumab in combination with sunitinib or pazopanib in advanced or metastatic renal cell carcinoma: the CheckMate 016 study. J Immunother Cancer 2018; 6:109. [PMID: 30348216 PMCID: PMC6196426 DOI: 10.1186/s40425-018-0420-0] [Citation(s) in RCA: 141] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 09/26/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Combination treatment with immune checkpoint inhibitors and antiangiogenic drugs has shown encouraging preliminary antitumor activity across various tumor types including advanced or metastatic renal cell carcinoma (aRCC). The open-label, parallel-cohort, dose-escalation, phase I CheckMate 016 study evaluated the efficacy and safety of nivolumab in combination with antiangiogenic tyrosine kinase inhibitors or ipilimumab. Long-term outcomes from this study for the combination of nivolumab plus sunitinib or pazopanib in aRCC are presented. METHODS Patients with aRCC received nivolumab plus either sunitinib (50 mg/day, 4 weeks on/2 weeks off; N + S) or pazopanib (800 mg/day; N + P) until progression/unacceptable toxicity. The nivolumab starting dose was 2 mg/kg every 3 weeks, with planned escalation to 5 mg/kg every 3 weeks. Primary endpoints were safety and tolerability; antitumor activity was a secondary endpoint. RESULTS Arm N + S enrolled 33 patients, 19 of whom were treatment-naïve; this arm advanced to the expansion phase. Median follow-up was 50.0 months. Patients experienced high frequencies of adverse events (AEs) including treatment-related AEs (100%), grade 3/4 treatment-related AEs (82%), and treatment-related AEs leading to discontinuation (39%). Investigator-assessed objective response rate (ORR) was 55% (18/33) and median progression-free survival (PFS) was 12.7 months. Median overall survival (OS) was not reached. Arm N + P enrolled 20 patients, all had ≥1 prior systemic therapy; this arm was closed due to dose-limiting toxicities and did not proceed to expansion. Median follow-up was 27.1 months. Patients treated with N + P experienced high frequencies of AEs including treatment-related AEs (100%), grade 3/4 treatment-related AEs (70%), and treatment-related AEs leading to discontinuation (25%). Investigator-assessed ORR was 45% (9/20) and median PFS was 7.2 months. Median OS was 27.9 months. CONCLUSIONS The addition of standard doses of sunitinib or pazopanib to nivolumab resulted in a high incidence of high-grade toxicities limiting future development of either combination regimen. While there was no adverse impact on response and the OS outcome was notable, the findings suggest that the success of combination regimens based on immune checkpoint inhibitors and antiangiogenic drugs may be dependent on careful selection of the antiangiogenic component and dose. TRIAL REGISTRATION Clinicaltrials.gov identifier: NCT01472081 . Registered 16 November 2011.
Collapse
Affiliation(s)
- Asim Amin
- Immunotherapy program, Levine Cancer Institute, Carolinas HealthCare System, 1024 Morehead Medical Drive, Charlotte, NC 28204 USA
| | - Elizabeth R Plimack
- Division of Genitourinary Medical Oncology, Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA 19111 USA
| | - Marc S Ernstoff
- Division of Oncology, Department of Medicine, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14203 USA
| | - Lionel D Lewis
- Department of Medicine at The Geisel School of Medicine and The Norris Cotton Cancer Center at Dartmouth-Hitchcock Medical Center, Lebanon, NH 03756 USA
| | - Todd M Bauer
- Sarah Cannon Research Institute/Tennessee Oncology, PLLC, Nashville, TN 37203 USA
| | - David F McDermott
- Department of Medicine, Beth Israel Deaconess Medical Center, Dana-Farber/Harvard Cancer Center, Boston, MA 02215 USA
| | - Michael Carducci
- Department of Oncology, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21287 USA
| | | | - Brian I Rini
- Lerner College of Medicine, Department of Hematology and Oncology, Cleveland Clinic Taussig Cancer Institute, Cleveland, OH 44195 USA
| | - Daniel Y C Heng
- Department of Oncology, Tom Baker Cancer Center, University of Calgary, Calgary, AB T2N 4N2 Canada
| | - Jennifer Knox
- Cancer Clinical Research Unit (CCRU), Princess Margaret Cancer Centre, Toronto, ON M5G 1Z5 Canada
| | - Martin H Voss
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065 USA
| | - Jennifer Spratlin
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB T6G 1Z2 Canada
| | - Elmer Berghorn
- Oncology - Global Clinical Research, Bristol-Myers Squibb, Princeton, NJ 08541 USA
| | - Lingfeng Yang
- Oncology - Global Clinical Research, Bristol-Myers Squibb, Princeton, NJ 08541 USA
| | - Hans J Hammers
- Department of Internal Medicine, UT Southwestern – Kidney Cancer Program, Dallas, TX 75390 USA
| |
Collapse
|
175
|
Kamal Y, Cheng C, Frost HR, Amos CI. Predictors of disease aggressiveness influence outcome from immunotherapy treatment in renal clear cell carcinoma. Oncoimmunology 2018; 8:e1500106. [PMID: 30546942 PMCID: PMC6287778 DOI: 10.1080/2162402x.2018.1500106] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 07/02/2018] [Accepted: 07/07/2018] [Indexed: 12/12/2022] Open
Abstract
Renal clear cell carcinoma (RCC) is the most common type of kidney cancer and has a high propensity for metastasis. While treatment with immune checkpoint inhibitors, such as anti-PD-1, have shown modest improvements in survival for RCC, it is difficult to identify responders from non-responders. Attempts to elucidate the mechanisms associated with differential response to checkpoint inhibitors have been limited by small sample size making it difficult to detect meaningful associations. We utilized existing large datasets from The Cancer Genome Atlas (TCGA) to first find predictors of disease aggressiveness in the tumor microenvironment (TME) and hypothesized that these same predictors may influence response to immunotherapy. We found primary metastatic (M1-stage IV) tumors exhibit high immune infiltration, and high TP53-inactivation induced senescence activity compared to non-metastatic (M0-Stage I/II) tumors. Moreover, some TME features inferred from deconvolution algorithms, which differ between M0 and M1 tumors, also influence overall survival. A focused analysis identified interactions between tumor TP53-inactivation induced senescence activity and expression of inflammatory molecules in pre-treatment RCC tumors, which predict both change in tumor size and response to checkpoint blockade therapy. We also noted frequency of inactivating mutations in the protein polybromo-1 (PBRM1) gene was found to be negatively associated with TP53-inactivation induced senescence enrichment. Our findings suggest a mechanism by which tumor TP53-inactivation induced senescence can modulate the TME and thereby influence outcome from checkpoint blockade therapy.
Collapse
Affiliation(s)
- Yasmin Kamal
- Department of Biomedical Data Sciences, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- Quantitative Biomedical Sciences, Baylor College of Medicine, Houston, TX, USA
| | - Chao Cheng
- Department of Biomedical Data Sciences, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- Quantitative Biomedical Sciences, Baylor College of Medicine, Houston, TX, USA
| | - H. Robert Frost
- Department of Biomedical Data Sciences, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- Quantitative Biomedical Sciences, Baylor College of Medicine, Houston, TX, USA
| | - Christopher I. Amos
- Department of Biomedical Data Sciences, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- Quantitative Biomedical Sciences, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
176
|
Gevaert T, Montironi R, Lopez-Beltran A, Van Leenders G, Allory Y, De Ridder D, Claessens F, Kockx M, Akand M, Joniau S, Netto G, Libbrecht L. Genito-urinary genomics and emerging biomarkers for immunomodulatory cancer treatment. Semin Cancer Biol 2018; 52:216-227. [DOI: 10.1016/j.semcancer.2017.10.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 10/04/2017] [Accepted: 10/06/2017] [Indexed: 01/01/2023]
|
177
|
Zahoor H, Pavicic PG, Przybycin C, Ko J, Stephens L, Radivoyevitch T, Jia X, Diaz-Montero CM, Finke J, Rayman PA, Gilligan TD, Grivas P, Ornstein M, Garcia JA, Rini BI. Evaluation of T cell infiltration in matched biopsy and nephrectomy samples in renal cell carcinoma. Medicine (Baltimore) 2018; 97:e12344. [PMID: 30212988 PMCID: PMC6156035 DOI: 10.1097/md.0000000000012344] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
T cell infiltration in tumors has been investigated as a biomarker of response to checkpoint inhibitors. Neo-adjuvant studies in renal cell carcinoma (RCC) may provide a unique opportunity to compare T cell infiltration in a pretreatment renal mass biopsy to a posttreatment nephrectomy specimen, and thus evaluate the effects of immune checkpoint inhibitors. However, there are no data regarding the association of T cell infiltration in matched biopsy and nephrectomy samples without intervening treatment. Understanding this association will inform investigation of this potential biomarker in future studies.Matched biopsy and nephrectomy samples (without intervening systemic therapy) were identified from patients with nonmetastatic RCC. Selected tissue sections from biopsy and nephrectomy samples were reviewed and marked for intratumoral lymphocytes by a pathologist. Immunohistochemistry (IHC) was utilized to stain for T cell markers (CD3, CD4, and CD8). Intratumoral staining was then quantified in the tissue sections as counts per total tumor area surveyed. Spearman correlation (r) was used to measure associations.Thirty matched pairs were investigated. The median interval between biopsy and nephrectomy was 2.8 (0.2-87.7) months. Clear cell was the most common histology (29/30; 97%). There was a statistically significant positive correlation between the frequency of CD3 and CD8 T cells between matched biopsy and nephrectomy samples (r = 0.39; P = .036 and r = 0.38; P = .041, respectively).The frequencies of CD8+ T cells in matched biopsy and nephrectomy samples in RCC in the absence of intervening treatment have been characterized and show a positive correlation between matched biopsy and nephrectomy samples.
Collapse
Affiliation(s)
| | | | | | - Jennifer Ko
- Robert J. Tomsich Pathology and Laboratory Medicine Institute
| | - Lisa Stephens
- Robert J. Tomsich Pathology and Laboratory Medicine Institute
| | - Tomas Radivoyevitch
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Xuefei Jia
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | | | - James Finke
- Department of Immunology, Lerner Research Institute
| | | | | | | | | | | | | |
Collapse
|
178
|
A comprehensive review of the role of immune checkpoint inhibitors in brain metastasis of renal cell carcinoma origin. Crit Rev Oncol Hematol 2018; 130:60-69. [PMID: 30196913 DOI: 10.1016/j.critrevonc.2018.08.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 07/15/2018] [Accepted: 08/01/2018] [Indexed: 02/06/2023] Open
Abstract
The contribution of renal cell carcinoma (RCC) to brain metastases (BM) reaches 7-13%. These patients have limited survival with local control and targeted therapies. Immune checkpoint inhibitors (ICI) revolutionized the treatment landscape of RCC but commonly excluded BM patients from their pivotal trials. The daily clinical practice often imposes the use of ICI in RCC patients with BM in view of the promising survival times and durations of response. Only small prospective trials have included BM patients but rarely reported on the efficacy or safety of ICI in this subgroup. The available data is limited to small retrospective and prospective series that have shown comparable efficacy to that of the pivotal trials. In this review, we will discuss the biological rationale and potential concerns for the use of ICI in BM RCC. Furthermore, we will summarize BM subgroup data from the prospective and retrospective series of ICI in RCC as well as the use of cranial radiation and ICI.
Collapse
|
179
|
Büttner F, Winter S, Rausch S, Hennenlotter J, Kruck S, Stenzl A, Scharpf M, Fend F, Agaimy A, Hartmann A, Bedke J, Schwab M, Schaeffeler E. Clinical utility of the S3-score for molecular prediction of outcome in non-metastatic and metastatic clear cell renal cell carcinoma. BMC Med 2018; 16:108. [PMID: 29973214 PMCID: PMC6033218 DOI: 10.1186/s12916-018-1088-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 06/01/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Stratification of cancer patients to identify those with worse prognosis is increasingly important. Through in silico analyses, we recently developed a gene expression-based prognostic score (S3-score) for clear cell renal cell carcinoma (ccRCC), using the cell type-specific expression of 97 genes within the human nephron. Herein, we verified the score using whole-transcriptome data of independent cohorts and extend its application for patients with metastatic disease receiving tyrosine kinase inhibitor treatment. Finally, we sought to improve the signature for clinical application using qRT-PCR. METHODS A 97 gene-based S3-score (S397) was evaluated in a set of 52 primary non-metastatic and metastatic ccRCC patients as well as in 53 primary metastatic tumors of sunitinib-treated patients. Gene expression data of The Cancer Genome Atlas (n = 463) was used for platform transfer and development of a simplified qRT-PCR-based 15-gene S3-score (S315). This S315-score was validated in 108 metastatic and non-metastatic ccRCC patients and ccRCC-derived metastases including in part several regions from one metastasis. Univariate and multivariate Cox regression stratified by T, N, M, and G were performed with cancer-specific and progression-free survival as primary endpoints. RESULTS The S397-score was significantly associated with cancer-specific survival (CSS) in 52 ccRCC patients (HR 2.9, 95% Cl 1.0-8.0, PLog-rank = 3.3 × 10-2) as well as progression-free survival in sunitinib-treated patients (2.1, 1.1-4.2, PLog-rank = 2.2 × 10-2). The qRT-PCR based S315-score performed similarly to the S397-score, and was significantly associated with CSS in our extended cohort of 108 patients (5.0, 2.1-11.7, PLog-rank = 5.1 × 10-5) including metastatic (9.3, 1.8-50.0, PLog-rank = 2.3 × 10-3) and non-metastatic patients (4.4, 1.2-16.3, PLog-rank = 1.6 × 10-2), even in multivariate Cox regression, including clinicopathological parameters (7.3, 2.5-21.5, PWald = 3.3 × 10-4). Matched primary tumors and metastases revealed similar S315-scores, thus allowing prediction of outcome from metastatic tissue. The molecular-based qRT-PCR S315-score significantly improved prediction of CSS by the established clinicopathological-based SSIGN score (P = 1.6 × 10-3). CONCLUSION The S3-score offers a new clinical avenue for ccRCC risk stratification in the non-metastatic, metastatic, and sunitinib-treated setting.
Collapse
Affiliation(s)
- Florian Büttner
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Auerbachstrasse 112, 70376, Stuttgart, Germany.,University of Tuebingen, Tuebingen, Germany
| | - Stefan Winter
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Auerbachstrasse 112, 70376, Stuttgart, Germany.,University of Tuebingen, Tuebingen, Germany
| | - Steffen Rausch
- Department of Urology, University Hospital Tuebingen, Tuebingen, Germany
| | - Jörg Hennenlotter
- Department of Urology, University Hospital Tuebingen, Tuebingen, Germany
| | - Stephan Kruck
- Department of Urology, University Hospital Tuebingen, Tuebingen, Germany
| | - Arnulf Stenzl
- Department of Urology, University Hospital Tuebingen, Tuebingen, Germany
| | - Marcus Scharpf
- Institute of Pathology and Neuropathology, University Hospital Tuebingen, Tuebingen, Germany
| | - Falko Fend
- Institute of Pathology and Neuropathology, University Hospital Tuebingen, Tuebingen, Germany
| | - Abbas Agaimy
- Institute of Pathology, Friedrich-Alexander University Erlangen-Nuernberg, University Hospital Erlangen-Nuernberg, Erlangen, Germany
| | - Arndt Hartmann
- Institute of Pathology, Friedrich-Alexander University Erlangen-Nuernberg, University Hospital Erlangen-Nuernberg, Erlangen, Germany
| | - Jens Bedke
- Department of Urology, University Hospital Tuebingen, Tuebingen, Germany.,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Matthias Schwab
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Auerbachstrasse 112, 70376, Stuttgart, Germany. .,University of Tuebingen, Tuebingen, Germany. .,German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany. .,Department of Clinical Pharmacology, University Hospital Tuebingen, Tuebingen, Germany. .,Department of Pharmacy and Biochemistry, University of Tuebingen, Tuebingen, Germany.
| | - Elke Schaeffeler
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Auerbachstrasse 112, 70376, Stuttgart, Germany. .,University of Tuebingen, Tuebingen, Germany.
| |
Collapse
|
180
|
Ferro S, Huber V, Rivoltini L. Mechanisms of tumor immunotherapy, with a focus on thoracic cancers. J Thorac Dis 2018; 10:4619-4631. [PMID: 30174914 DOI: 10.21037/jtd.2018.07.30] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
If immunotherapy is currently considered as a valid treatment strategy in oncology, the concept that cancer could be cured by the mere manipulation of the immune system was almost inconceivable until few years ago, particularly in lung cancer. The use of immune checkpoint inhibitors has instead demonstrated to mediate significant long-term disease control so to rapidly enter clinical practice and represent the basis for most of the combination approaches under development. In light of the revolutionary results achieved through the pivotal clinical trials and the large expectations about the possibility to further improve clinical benefit and discover novel therapeutic targets, it is becoming nowadays mandatory to increase our knowledge on the basics of immunology in lung cancer. Defining the pathways that rule the interactions between tumor and immune cells and the requirements to achieve full-fledged immune responses able to mediate meaningful antitumor activity are present goals of the research ongoing worldwide. This knowledge would not only foster a more scientifically-based clinical development of novel drugs and combinations, but also provide valid biomarkers for patient selection and monitoring. In the present review we will address the available information about the immunological features of lung cancer, the backgrounds to the use of immunotherapeutics, the possible mechanisms underlying resistance and the strategies to improve immune-mediated tumor control. In doing this, we will be following the path traced in melanoma, the tumor histotype that taught us most of what we know about cancer immunotherapy.
Collapse
Affiliation(s)
- Simona Ferro
- Unit of Immunotherapy of Human Tumors, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Veronica Huber
- Unit of Immunotherapy of Human Tumors, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Licia Rivoltini
- Unit of Immunotherapy of Human Tumors, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
181
|
The absolute lymphocyte count can predict the overall survival of patients with non-small cell lung cancer on nivolumab: a clinical study. Clin Transl Oncol 2018; 21:206-212. [PMID: 29948974 DOI: 10.1007/s12094-018-1908-2] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 06/04/2018] [Indexed: 10/14/2022]
Abstract
INTRODUCTION The neutrophil-to-lymphocyte (ANC/ALC) ratio is associated with worse prognosis in patients with NSCLC on immunotherapies, but the role of ALC remains unclear. The previous radiation therapy causes lymphopenia, and given approaches of combining radiation with immunotherapies, it is critical to better understand the impact of peripheral lymphocytes. PATIENTS AND METHODS We evaluated retrospectively 22 patients with advanced NSCLC treated with nivolumab at Boston Medical Center from January 2014 to September 2016 and correlated the peripheral blood counts with the overall survival (OS) and overall time on treatment. We assessed the effect of the previous radiation on peripheral blood counts and clinical outcomes. RESULTS Baseline ALC and ANC/ALC ratios are positively and negatively correlated, respectively, with the OS on nivolumab. The ALC and ALC/WBC ratios at 6 weeks on treatment are positively associated with the OS. Kaplan-Meier analysis at baseline and at 6 weeks showed significantly increased OS in the group of patients with the highest ALC. The previous radiation therapy was positively correlated with the ANC and negatively correlated with the ALC/WBC ratio at 8 weeks after the initiation of nivolumab. CONCLUSION Our finding that ALC at baseline and at 6 weeks on treatment is positively correlated with the OS provides an easily obtained predictive marker. Our result that the previous radiation is associated with higher ANC and lower ALC during treatment supports that the combination of radiation therapy with immunotherapy should be carefully applied and potentially peripheral blood counts can be utilized to stratify patients for this approach.
Collapse
|
182
|
Zarrabi K, Wu S. An evaluation of nivolumab for the treatment of metastatic renal cell carcinoma. Expert Opin Biol Ther 2018; 18:695-705. [PMID: 29782188 DOI: 10.1080/14712598.2018.1478962] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION The treatment paradigm for metastatic renal cell carcinoma (mRCC) has undergone a revolution with the rapid market approval of multiple agents over a three-year period. The immunogenicity of renal cell carcinoma (RCC) provided the biological rationale to assess the clinical efficacy of nivolumab, an immune checkpoint inhibitor. Nivolumab is approved for second-line treatment after failure of angiogenesis-targeted therapy and in combination therapy with ipilimumab for previously untreated intermediate- or poor-risk advanced RCC. Areas covered: The authors review the preclinical and clinical data supporting nivolumab employment in mRCC. Discussion of the underlying mechanisms of immunotherapy, data on objective responses, safety and tolerability, regulatory affairs, and future directions of nivolumab therapy are highlighted. Expert opinion: Nivolumab serves as a proof-of-principle of the efficacy of immunotherapy with checkpoint inhibition in mRCC. Nivolumab may be considered the leading monotherapy in the second-line setting for patients with low tumor volume considering its risks and benefits. Nivolumab was recently approved in the first-line setting as part of combination therapy with another immune modulator. Moreover, nivolumab use may offer clinicians the option for treatment beyond progression as emerging data has indicated possible overall survival benefits in this setting. Ongoing clinical studies may result in nivolumab use in the first-line setting, as monotherapy or in combination therapy with antiangiogenesis agents.
Collapse
Affiliation(s)
- Kevin Zarrabi
- a Department of Medicine , Stony Brook University Hospital , Stony Brook , NY , USA
| | - Shenhong Wu
- b Division of Hematology/Oncology, Department of Medicine , Northport VA Medical Center , Northport , NY , USA
| |
Collapse
|
183
|
Lyu C, Li W, Liu S, Gao S, Zhang H, Hao L, Yu H, Wei W, Song J, Yang Y, Wang C, Zhang Z, Wang N. Systematic review on the efficacy and safety of immune checkpoint inhibition in renal cell carcinoma. Future Oncol 2018; 14:2207-2221. [PMID: 29726696 DOI: 10.2217/fon-2018-0193] [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
To perform a systematic review of the relevant literature about clinical trials on efficacy and safety of immune checkpoint inhibition, whether it is used alone, in combination or with other targeted therapies in patients with advanced and metastatic renal cell carcinoma (RCC), two team members reviewed the abstracts and selected pertinent articles from the relevant databases. A narrative review of randomized controlled trials was performed and seven randomized controlled trials were identified in this systematic review. In treatment of RCC, nivolumab has superior efficacy and safety compared with second-line everolimus. Combination strategies, especially those combined with anti-VEGF agents presents better efficacy but worse outcomes in term of safety than monotherapy and conventional treatment and might guide treatment choice for patients with RCC.
Collapse
Affiliation(s)
- Chen Lyu
- College of Animal Science, Jilin University, Changchun, PR China
| | - Wenyue Li
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, PR China
| | - Songcai Liu
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, PR China.,Five-Star Animal Health Pharmaceutical Factory of Jilin Province, Changchun, PR China
| | - Shuang Gao
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, PR China
| | - Hong Zhang
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, PR China
| | - Linlin Hao
- College of Animal Science, Jilin University, Changchun, PR China
| | - Hao Yu
- College of Animal Science, Jilin University, Changchun, PR China
| | - Wenzhen Wei
- College of Animal Science, Jilin University, Changchun, PR China
| | - Jie Song
- College of Animal Science, Jilin University, Changchun, PR China
| | - Yaoyao Yang
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, PR China
| | - Chunmeng Wang
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, PR China
| | - Zhimin Zhang
- College of Animal Science, Jilin University, Changchun, PR China
| | - Nan Wang
- China Institute of Veterinary Drug Control, Beijing, PR China
| |
Collapse
|
184
|
Bedke J, Grimm MO, Grünwald V. Collection of real-world data on nivolumab's effectiveness in renal cell carcinoma: rationale for an observational study. Future Oncol 2018; 14:1023-1034. [DOI: 10.2217/fon-2017-0637] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Renal cell carcinoma (RCC) represents the seventh (men) respectively tenth (women) most frequent cancer in western countries. After one or more lines of VEGF-targeted therapy, immunotherapy with nivolumab is strongly recommended in patients with metastatic RCC. Nivolumab is the first, and so far, only approved PD-1 immune checkpoint inhibitor to demonstrate a gain in overall survival in RCC. We describe herein design and rationale of trial CA209653 (‘NIS NORA’), a prospective, noninterventional cohort study investigating the effectiveness of nivolumab. This systematic collection of real-world effectiveness data will recruit 323 patients with advanced RCC to provide a precise estimate for overall survival over a 5-year follow-up period (Trial registration: NCT02940639).
Collapse
Affiliation(s)
- Jens Bedke
- Department of Urology, University Hospital Tübingen, Tübingen, Germany
| | | | - Viktor Grünwald
- Department of Hematology, Hemostasis, Oncology & Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| |
Collapse
|
185
|
Soule E, Bandyk M, Matteo J. Percutaneous ablative cryoimmunotherapy for micrometastaic abscopal effect: No complications. Cryobiology 2018; 82:22-26. [PMID: 29680230 DOI: 10.1016/j.cryobiol.2018.04.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 04/16/2018] [Accepted: 04/18/2018] [Indexed: 01/05/2023]
Abstract
Recent studies have shown efficacy of nivolumab, a monoclonal antibody specific for an immunoregulatory protein termed programmed death 1 (PD-1), against metastatic renal cell carcinoma. PD-1 is a transmembrane protein expressed on T cells that suppresses activation upon binding to its ligands, which may be secreted paraneoplastically by various cancers. Inhibition of PD-1 signaling via nivolumab may sensitize tumor-specific T lymphocytes in the immunosuppressive tumor microenvironment. Systemic elimination of micrometastases requires robust activation and proliferation of tumor antigen stimulated T cells. Cryoablation lyses tumor cell membranes but leaves intracellular tumor antigens intact for recognition by effectors systemically, while eliminating permissive T cell subtypes locally. This single case report describes CT-guided percutaneous cryoablation of a metastatic renal cell carcinoma with local administration of nivolumab to simultaneously debulk the primary tumor, sensitize effector T cells against tumor antigens, and augment the systemic immune response elicited against established metastases. One month follow up PET scan revealed decreased uptake in the two smaller metastatic bone lesions with the smallest lesion completely eliminated. The largest metastatic bone lesion was slightly decreased in size and exhibited slightly increased uptake. The patient's presenting complaint of hip pain was abrogated, allowing her to resume independent ambulation. CT-guided percutaneous cryoablation is minimally invasive and preserves tumor antigens, which are subsequently presented to tumor-specific T cells; their differentiation into cytotoxic T cells may be guided and their proliferation may be augmented by local administration of immunostimulatory pharmacotherapy at the time of the procedure.
Collapse
Affiliation(s)
- Erik Soule
- Department of Interventional Radiology, UF Health Jacksonville, University of Florida, 655 W 8th, St., Jacksonville, FL, 32209, USA.
| | - Mark Bandyk
- Department of Urology, UF Health Jacksonville, University of Florida, 655 W 8th, St., Jacksonville, FL, 32209, USA.
| | - Jerry Matteo
- Department of Interventional Radiology, UF Health Jacksonville, University of Florida, 655 W 8th, St., Jacksonville, FL, 32209, USA.
| |
Collapse
|
186
|
Frelau A, Pracht M, Le Sourd S, Lespagnol A, Corre R, Ménard C, Tarte K, Mosser J, Edeline J. WITHDRAWN: Biomarqueurs prédictifs de réponse aux inhibiteurs de points de contrôle immuns. Bull Cancer 2018:S0007-4551(17)30384-3. [PMID: 29525053 DOI: 10.1016/j.bulcan.2017.10.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 10/19/2017] [Accepted: 10/23/2017] [Indexed: 11/19/2022]
Affiliation(s)
- Alexandra Frelau
- CLCC Eugène-Marquis UNICANCER, oncologie médicale, avenue de la Bataille-Flandres-Dunkerque, 35042 Rennes cedex, France.
| | - Marc Pracht
- CLCC Eugène-Marquis UNICANCER, oncologie médicale, avenue de la Bataille-Flandres-Dunkerque, 35042 Rennes cedex, France
| | - Samuel Le Sourd
- CLCC Eugène-Marquis UNICANCER, oncologie médicale, avenue de la Bataille-Flandres-Dunkerque, 35042 Rennes cedex, France
| | - Alexandra Lespagnol
- CHU de Rennes, laboratoire de génétique somatique des cancers, 2, rue Henri-Le-Guilloux, 35033 Rennes cedex 9, France
| | - Romain Corre
- CHU de Rennes, service de pneumologie, 2, rue Henri-Le-Guilloux, 35033 Rennes cedex 9, France
| | - Cédric Ménard
- CHU de Rennes, Inserm U197, laboratoire d'immunologie, thérapie cellulaire et hématopoïèse, 2, rue Henri-Le-Guilloux, 35033 Rennes cedex 9, France
| | - Karin Tarte
- CHU de Rennes, Inserm U197, laboratoire d'immunologie, thérapie cellulaire et hématopoïèse, 2, rue Henri-Le-Guilloux, 35033 Rennes cedex 9, France
| | - Jean Mosser
- CHU de Rennes, laboratoire de génétique somatique des cancers, 2, rue Henri-Le-Guilloux, 35033 Rennes cedex 9, France; CHU de Rennes, institut de génétique et développement, CNRS-UR1, IGRD UMR 6290, 2, rue Henri-Le-Guilloux, 35033 Rennes cedex 9, France
| | - Julien Edeline
- CLCC Eugène-Marquis UNICANCER, oncologie médicale, avenue de la Bataille-Flandres-Dunkerque, 35042 Rennes cedex, France; CHU de Rennes, UMR991, unité de recherche foie, métabolisme et cancer, 2, rue Henri-Le-Guilloux, 35033 Rennes cedex 9, France
| |
Collapse
|
187
|
Atkins MB, Plimack ER, Puzanov I, Fishman MN, McDermott DF, Cho DC, Vaishampayan U, George S, Olencki TE, Tarazi JC, Rosbrook B, Fernandez KC, Lechuga M, Choueiri TK. Axitinib in combination with pembrolizumab in patients with advanced renal cell cancer: a non-randomised, open-label, dose-finding, and dose-expansion phase 1b trial. Lancet Oncol 2018; 19:405-415. [PMID: 29439857 PMCID: PMC6860026 DOI: 10.1016/s1470-2045(18)30081-0] [Citation(s) in RCA: 286] [Impact Index Per Article: 47.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Revised: 12/20/2017] [Accepted: 12/20/2017] [Indexed: 12/26/2022]
Abstract
BACKGROUND Previous studies combining PD-1 checkpoint inhibitors with tyrosine kinase inhibitors of the VEGF pathway have been characterised by excess toxicity, precluding further development. We hypothesised that axitinib, a more selective VEGF inhibitor than others previously tested, could be combined safely with pembrolizumab (anti-PD-1) and yield antitumour activity in patients with treatment-naive advanced renal cell carcinoma. METHODS In this ongoing, open-label, phase 1b study, which was done at ten centres in the USA, we enrolled patients aged 18 years or older who had advanced renal cell carcinoma (predominantly clear cell subtype) with their primary tumour resected, and at least one measureable lesion, Eastern Cooperative Oncology Group performance status 0-1, controlled hypertension, and no previous systemic therapy for renal cell carcinoma. Eligible patients received axitinib plus pembrolizumab in a dose-finding phase to estimate the maximum tolerated dose, and additional patients were enrolled into a dose-expansion phase to further establish safety and determine preliminary efficacy. Axitinib 5 mg was administered orally twice per day with pembrolizumab 2 mg/kg given intravenously every 3 weeks. We assessed safety in all patients who received at least one dose of axitinib or pembrolizumab; antitumour activity was assessed in all patients who received study treatment and had an adequate baseline tumour assessment. The primary endpoint was investigator-assessed dose-limiting toxicity during the first two cycles (6 weeks) to estimate the maximum tolerated dose and recommended phase 2 dose. This study is registered with ClinicalTrials.gov, number NCT02133742. FINDINGS Between Sept 23, 2014, and March 25, 2015, we enrolled 11 patients with previously untreated advanced renal cell carcinoma to the dose-finding phase and between June 3, 2015, and Oct 13, 2015, we enrolled 41 patients to the dose-expansion phase. All 52 patients were analysed together. No unexpected toxicities were observed. Three dose-limiting toxicities were reported in the 11 patients treated during the 6-week observation period (dose-finding phase): one patient had a transient ischaemic attack and two patients were only able to complete less than 75% of the planned axitinib dose because of treatment-related toxicity. At the data cutoff date (March 31, 2017), 25 (48%) patients were still receiving study treatment. Grade 3 or worse treatment-related adverse events occurred in 34 (65%) patients; the most common included hypertension (n=12 [23%]), diarrhoea (n=5 [10%]), fatigue (n=5 [10%]), and increased alanine aminotransferase concentration (n=4 [8%]). The most common potentially immune-related adverse events (probably related to pembrolizumab) included diarrhoea (n=15 [29%]), increased alanine aminotransferase concentration (n=9 [17%]) or aspartate aminotransferase concentration (n=7 [13%]), hypothyroidism (n=7 [13%]), and fatigue (n=6 [12%]). 28 (54%) patients had treatment-related serious adverse events. At data cutoff, 38 (73%; 95% CI 59·0-84·4) patients achieved an objective response (complete or partial response). INTERPRETATION The treatment combination of axitinib plus pembrolizumab is tolerable and shows promising antitumour activity in patients with treatment-naive advanced renal cell carcinoma. Whether or not the combination works better than a sequence of VEGF pathway inhibition followed by an anti-PD-1 therapy awaits the completion of a phase 3 trial comparing axitinib plus pembrolizumab with sunitinib monotherapy (NCT02853331). FUNDING Pfizer Inc.
Collapse
MESH Headings
- Aged
- Angiogenesis Inhibitors/administration & dosage
- Angiogenesis Inhibitors/adverse effects
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/adverse effects
- Antineoplastic Agents, Immunological/administration & dosage
- Antineoplastic Agents, Immunological/adverse effects
- Antineoplastic Combined Chemotherapy Protocols/administration & dosage
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Axitinib/administration & dosage
- Axitinib/adverse effects
- Carcinoma, Renal Cell/drug therapy
- Carcinoma, Renal Cell/mortality
- Carcinoma, Renal Cell/pathology
- Carcinoma, Renal Cell/surgery
- Chemotherapy, Adjuvant
- Dose-Response Relationship, Drug
- Drug Dosage Calculations
- Female
- Humans
- Kidney Neoplasms/drug therapy
- Kidney Neoplasms/mortality
- Kidney Neoplasms/pathology
- Kidney Neoplasms/surgery
- Male
- Middle Aged
- Nephrectomy
- Time Factors
- Treatment Outcome
- United States
Collapse
Affiliation(s)
- Michael B Atkins
- Georgetown-Lombardi Comprehensive Cancer Center, Washington, DC, USA.
| | | | - Igor Puzanov
- Vanderbilt University Medical Center, Nashville, TN, USA; Roswell Park Cancer Institute, Buffalo, NY, USA
| | | | | | - Daniel C Cho
- New York University Langone Medical Center, New York, NY, USA
| | | | - Saby George
- Roswell Park Cancer Institute, Buffalo, NY, USA
| | | | | | | | | | | | | |
Collapse
|
188
|
Guo Y, Walsh AM, Canavan M, Wechalekar MD, Cole S, Yin X, Scott B, Loza M, Orr C, McGarry T, Bombardieri M, Humby F, Proudman SM, Pitzalis C, Smith MD, Friedman JR, Anderson I, Madakamutil L, Veale DJ, Fearon U, Nagpal S. Immune checkpoint inhibitor PD-1 pathway is down-regulated in synovium at various stages of rheumatoid arthritis disease progression. PLoS One 2018; 13:e0192704. [PMID: 29489833 PMCID: PMC5831027 DOI: 10.1371/journal.pone.0192704] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 01/29/2018] [Indexed: 12/30/2022] Open
Abstract
Immune checkpoint blockade with therapeutic anti-cytotoxic T lymphocyte-associated antigen (CTLA)-4 (Ipilimumab) and anti-programmed death (PD)-1 (Nivolumab and Pembrolizumab) antibodies alone or in combination has shown remarkable efficacy in multiple cancer types, concomitant with immune-related adverse events, including arthralgia and inflammatory arthritis (IA) in some patients. Herein, using Nivolumab (anti-PD-1 antagonist)-responsive genes along with transcriptomics of synovial tissue from multiple stages of rheumatoid arthritis (RA) disease progression, we have interrogated the activity status of PD-1 pathway during RA development. We demonstrate that the expression of PD-1 was increased in early and established RA synovial tissue compared to normal and OA synovium, whereas that of its ligands, programmed death ligand-1 (PD-L1) and PD-L2, was increased at all the stages of RA disease progression, namely arthralgia, IA/undifferentiated arthritis, early RA and established RA. Further, we show that RA patients expressed PD-1 on a majority of synovial tissue infiltrating CD4+ and CD8+ T cells. Moreover, enrichment of Nivolumab gene signature was observed in IA and RA, indicating that the PD-1 pathway was downregulated during RA disease progression. Furthermore, serum soluble (s) PD-1 levels were increased in autoantibody positive early RA patients. Interestingly, most of the early RA synovium tissue sections showed negative PD-L1 staining by immunohistochemistry. Therefore, downregulation in PD-1 inhibitory signaling in RA could be attributed to increased serum sPD-1 and decreased synovial tissue PD-L1 levels. Taken together, these data suggest that agonistic PD1 antibody-based therapeutics may show efficacy in RA treatment and interception.
Collapse
Affiliation(s)
- Yanxia Guo
- Immunology, Janssen Research, Pennsylvania, United States of America
| | - Alice M. Walsh
- Immunology, Janssen Research, Pennsylvania, United States of America
| | - Mary Canavan
- Molecular Rheumatology, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland
| | - Mihir D. Wechalekar
- Rheumatology Unit, Repatriation General Hospital and Flinders University, Adelaide, Australia
| | - Suzanne Cole
- Immunology, Janssen Research, Pennsylvania, United States of America
| | - Xuefeng Yin
- Immunology, Janssen Research, Pennsylvania, United States of America
| | - Brittney Scott
- Immunology, Janssen Research, Pennsylvania, United States of America
| | - Mathew Loza
- Immunology, Janssen Research, Pennsylvania, United States of America
| | - Carl Orr
- Centre for Arthritis and Rheumatic Diseases, St. Vincent's University Hospital, University College Dublin, Elm Park, Dublin, Ireland
| | - Trudy McGarry
- Molecular Rheumatology, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland
| | - Michele Bombardieri
- Queen Mary University of London, Charterhouse Square, London, United Kingdom
| | - Frances Humby
- Queen Mary University of London, Charterhouse Square, London, United Kingdom
| | - Susanna M. Proudman
- Rheumatology Unit, Royal Adelaide Hospital, Adelaide, Australia and Discipline of Medicine, University of Adelaide, Adelaide, Australia
| | - Costantino Pitzalis
- Queen Mary University of London, Charterhouse Square, London, United Kingdom
| | - Malcolm D. Smith
- Rheumatology Unit, Repatriation General Hospital and Flinders University, Adelaide, Australia
| | | | - Ian Anderson
- Immunology, Janssen Research, Pennsylvania, United States of America
| | - Loui Madakamutil
- Immunology, Janssen Research, Pennsylvania, United States of America
| | - Douglas J. Veale
- Centre for Arthritis and Rheumatic Diseases, St. Vincent's University Hospital, University College Dublin, Elm Park, Dublin, Ireland
| | - Ursula Fearon
- Molecular Rheumatology, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland
- * E-mail: (SN); (UF)
| | - Sunil Nagpal
- Immunology, Janssen Research, Pennsylvania, United States of America
- * E-mail: (SN); (UF)
| |
Collapse
|
189
|
Arasaratnam M, Gurney H. Nivolumab in the treatment of advanced renal cell carcinoma. Future Oncol 2018; 14:1679-1689. [PMID: 29460635 DOI: 10.2217/fon-2017-0533] [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
Renal cell carcinoma (RCC) has typically been considered an immunogenic malignancy with responses seen to IL-2 and IFN-α. Response rates, however, were low and at the cost of considerable toxicity and as such, agents targeting angiogenesis have become the mainstay of treatment. Nivolumab is an immune checkpoint inhibitor targeting PD-1 thereby upregulating the host immune response against tumor cells. Nivolumab has emerged as a promising new therapy in advanced malignancies, and the first agent to show survival advantage in patients failing prior VEGFR-targeted therapy in metastatic RCC. This review summarizes the present evidence, toxicity profile, potential predictive biomarkers and promising future strategies with nivolumab in metastatic RCC.
Collapse
Affiliation(s)
- Malmaruha Arasaratnam
- Department of Medical Oncology, Crown Princess Mary Cancer Centre, Westmead Hospital, Sydney, Australia
| | - Howard Gurney
- Department of Medical Oncology, Crown Princess Mary Cancer Centre, Westmead Hospital, Sydney, Australia.,Macquarie University Clinic, Macquarie University Hospital, Sydney, Australia
| |
Collapse
|
190
|
Miao D, Margolis CA, Gao W, Voss MH, Li W, Martini DJ, Norton C, Bossé D, Wankowicz SM, Cullen D, Horak C, Wind-Rotolo M, Tracy A, Giannakis M, Hodi FS, Drake CG, Ball MW, Allaf ME, Snyder A, Hellmann MD, Ho T, Motzer RJ, Signoretti S, Kaelin WG, Choueiri TK, Van Allen EM. Genomic correlates of response to immune checkpoint therapies in clear cell renal cell carcinoma. Science 2018; 359:801-806. [PMID: 29301960 PMCID: PMC6035749 DOI: 10.1126/science.aan5951] [Citation(s) in RCA: 842] [Impact Index Per Article: 140.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 10/30/2017] [Accepted: 12/15/2017] [Indexed: 12/20/2022]
Abstract
Immune checkpoint inhibitors targeting the programmed cell death 1 receptor (PD-1) improve survival in a subset of patients with clear cell renal cell carcinoma (ccRCC). To identify genomic alterations in ccRCC that correlate with response to anti-PD-1 monotherapy, we performed whole-exome sequencing of metastatic ccRCC from 35 patients. We found that clinical benefit was associated with loss-of-function mutations in the PBRM1 gene (P = 0.012), which encodes a subunit of the PBAF switch-sucrose nonfermentable (SWI/SNF) chromatin remodeling complex. We confirmed this finding in an independent validation cohort of 63 ccRCC patients treated with PD-1 or PD-L1 (PD-1 ligand) blockade therapy alone or in combination with anti-CTLA-4 (cytotoxic T lymphocyte-associated protein 4) therapies (P = 0.0071). Gene-expression analysis of PBAF-deficient ccRCC cell lines and PBRM1-deficient tumors revealed altered transcriptional output in JAK-STAT (Janus kinase-signal transducers and activators of transcription), hypoxia, and immune signaling pathways. PBRM1 loss in ccRCC may alter global tumor-cell expression profiles to influence responsiveness to immune checkpoint therapy.
Collapse
Affiliation(s)
- Diana Miao
- 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
| | - Claire A Margolis
- 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
| | - Wenhua Gao
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Martin H Voss
- Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Weill Cornell Medical College, New York, NY 10065, USA
| | - Wei Li
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Dylan J Martini
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Craig Norton
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Dominick Bossé
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Stephanie M Wankowicz
- 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
| | - Dana Cullen
- Bristol-Myers Squibb, New York, NY 10154, USA
| | | | | | - Adam Tracy
- Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA 02142, USA
| | - Marios Giannakis
- 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
| | - Frank Stephen Hodi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | | | - Mark W Ball
- James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Mohamad E Allaf
- James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | | | - Matthew D Hellmann
- Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Weill Cornell Medical College, New York, NY 10065, USA
| | - Thai Ho
- Mayo Clinic, Scottsdale, AZ 85259, USA
| | - Robert J Motzer
- Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Weill Cornell Medical College, New York, NY 10065, USA
| | - Sabina Signoretti
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - William G Kaelin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Howard Hughes Medical Institute, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Toni K Choueiri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA.
| | - Eliezer M 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
|
191
|
Sanfrancesco JM, Cheng L. Complexity of the genomic landscape of renal cell carcinoma: Implications for targeted therapy and precision immuno-oncology. Crit Rev Oncol Hematol 2017; 119:23-28. [DOI: 10.1016/j.critrevonc.2017.09.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 09/05/2017] [Accepted: 09/20/2017] [Indexed: 12/29/2022] Open
|
192
|
Hoerner-Rieber J, Duma M, Blanck O, Hildebrandt G, Wittig A, Lohaus F, Flentje M, Mantel F, Krempien R, Eble MJ, Kahl KH, Boda-Heggemann J, Rieken S, Guckenberger M. Stereotactic body radiotherapy (SBRT) for pulmonary metastases from renal cell carcinoma-a multicenter analysis of the German working group "Stereotactic Radiotherapy". J Thorac Dis 2017; 9:4512-4522. [PMID: 29268521 DOI: 10.21037/jtd.2017.10.108] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Background Renal cell carcinoma (RCC) is traditionally considered to be radioresistant. Radiotherapy response rates are believed to improve with hypofractionated, high dose stereotactic body radiotherapy (SBRT). However, limited data exist regarding the role of SBRT in the treatment of pulmonary metastases. Methods The working group "Stereotactic Radiotherapy" of the German Society of Radiation Oncology analyzed its multi-institutional database of more than 700 patients who received SBRT for pulmonary metastases. Treatment was performed at 10 centers between 2001 and 2016. Patients with metastatic RCC were included in the study. Tumor characteristics, treatment details, and follow-up data including survival, local control (LC), distant metastases, and toxicity were evaluated. Results A total of 46 RCC patients treated with SBRT for 67 lung metastases were identified, who received a median total biologically effective dose (BEDiso) at planning target volume (PTV) isocenter of 117.0 Gy (range, 48.0-189.0 Gy). A median fractional dose of 20.8 Gy at isocenter (range, 6.0-37.9 Gy) was administered in a median number of 3 fractions (1-8 fractions). After a median follow-up time of 28.3 months for all patients, 1- and 3-year LC rates were 98.1% and 91.9%, with corresponding 1- and 3-year overall survival (OS) of 84.3% and 43.8%, respectively. Pulmonary metastases treated with BEDiso ≥130 Gy showed a trend for superior LC (P=0.054). OS was significantly improved in both uni- and multivariate analysis for patients with higher Karnofsky performance scale, lower maximum pulmonary metastasis diameter and lack of post-SBRT systemic therapy due to progression (P=0.014; P=0.049; P=0.006). Only mild acute and late toxicity was reported. Conclusions SBRT for pulmonary metastases from RCC was associated with low treatment-associated toxicity, promising survival, and excellent LC, especially in those patients receiving a BEDiso ≥130 Gy.
Collapse
Affiliation(s)
- Juliane Hoerner-Rieber
- Department of Radiation Oncology, University Hospital Heidelberg, Heidelberg, Germany.,National Center for Radiation Oncology (NCRO), Heidelberg Institute for Radiation Oncology (HIRO), Heidelberg, Germany
| | - Marciana Duma
- Department of Radiation Oncology, Technical University Munich, Germany
| | - Oliver Blanck
- Department of Radiation Oncology, UKSH Universitätsklinikum Schleswig-Holstein, Kiel, Germany
| | - Guido Hildebrandt
- Department of Radiation Oncology, University of Rostock, Rostock, Germany
| | - Andrea Wittig
- Department of Radiotherapy and Radiation Oncology, Philipps-University Marburg, University Hospital Giessen and Marburg, Germany
| | - Fabian Lohaus
- Department of Radiation Oncology, Medical Faculty and University Hospital C.G. Carus, Technical University Dresden, Germany.,German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) partner site Dresden, Germany.,OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Germany
| | - Michael Flentje
- Department of Radiation Oncology, University Hospital Wuerzburg, Germany
| | - Frederick Mantel
- Department of Radiation Oncology, University Hospital Wuerzburg, Germany
| | - Robert Krempien
- Department of Radiation Oncology, Helios Klinikum Berlin Buch, Berlin, Germany
| | - Michael J Eble
- Department of Radiation Oncology, University Hospital Aachen, Aachen, Germany
| | | | - Judit Boda-Heggemann
- Department of Radiation Oncology, University Medical Center Mannheim, University of Heidelberg, Germany
| | - Stefan Rieken
- Department of Radiation Oncology, University Hospital Heidelberg, Heidelberg, Germany.,National Center for Radiation Oncology (NCRO), Heidelberg Institute for Radiation Oncology (HIRO), Heidelberg, Germany
| | - Matthias Guckenberger
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Switzerland
| |
Collapse
|
193
|
Filley AC, Henriquez M, Dey M. Recurrent glioma clinical trial, CheckMate-143: the game is not over yet. Oncotarget 2017; 8:91779-91794. [PMID: 29207684 PMCID: PMC5710964 DOI: 10.18632/oncotarget.21586] [Citation(s) in RCA: 267] [Impact Index Per Article: 38.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 09/08/2017] [Indexed: 12/31/2022] Open
Abstract
Glioblastoma (GBM) is the most common, and aggressive, primary brain tumor in adults. With a median patient survival of less than two years, GBM represents one of the biggest therapeutic challenges of the modern era. Even with the best available treatment, recurrence rates are nearly 100% and therapeutic options at the time of relapse are extremely limited. Nivolumab, an anti-programmed cell death-1 (PD-1) monoclonal antibody, has provided significant clinical benefits in the treatment of various advanced cancers and represented a promising therapy for primary and recurrent GBM. CheckMate 143 (NCT 02017717) was the first large randomized clinical trial of PD pathway inhibition in the setting of GBM, including a comparison of nivolumab and the anti-VEGF antibody, bevacizumab, in the treatment of recurrent disease. However, preliminary results, recently announced in a WFNOS 2017 abstract, demonstrated a failure of nivolumab to prolong overall survival of patients with recurrent GBM, and this arm of the trial was prematurely closed. In this review, we discuss the basic concepts underlying the rational to target PD pathway in GBM, address implications of using immune checkpoint inhibitors in central nervous system malignancies, provide a rationale for possible reasons contributing to the failure of nivolumab to prolong survival in patients with recurrent disease, and analyze the future role of immune checkpoint inhibitors in the treatment of GBM.
Collapse
Affiliation(s)
- Anna C. Filley
- Department of Neurosurgery, Indiana University Purdue University Indianapolis, Indianapolis, Indiana, USA
| | - Mario Henriquez
- Department of Neurosurgery, Indiana University Purdue University Indianapolis, Indianapolis, Indiana, USA
| | - Mahua Dey
- Department of Neurosurgery, Indiana University Purdue University Indianapolis, Indianapolis, Indiana, USA
| |
Collapse
|
194
|
Ogawara D, Soda H, Iwasaki K, Suyama T, Taniguchi H, Fukuda Y, Mukae H. Remarkable response of nivolumab-refractory lung cancer to salvage chemotherapy. Thorac Cancer 2017; 9:175-180. [PMID: 29063735 PMCID: PMC5754301 DOI: 10.1111/1759-7714.12543] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 09/14/2017] [Accepted: 09/14/2017] [Indexed: 12/11/2022] Open
Abstract
Promising outcomes of salvage chemotherapy after nivolumab therapy have been reported; however, little is known about the detailed clinical and immunologic features in lung cancer patients in whom nivolumab is unsuccessful. We report two cases of nivolumab‐refractory lung cancer, in which chemotherapy resulted in rapid regression of the lung cancer. Upon initial diagnosis, the biopsy specimens showed PD‐ligand 1 (PD‐L1)‐expressing cancer cells, accompanied by tumor‐infiltrating lymphocytes with a favorable CD8/CD4 ratio. Immunosuppressive regulatory T cells and cells positive for TIM‐3 were also observed. Physicians should take caution in treating lung cancer patients after progression on nivolumab. Further studies with a large cohort are warranted to identify the patients that may benefit from salvage chemotherapy.
Collapse
Affiliation(s)
- Daiki Ogawara
- Department of Respiratory Medicine, Sasebo City General Hospital, Nagasaki, Japan
| | - Hiroshi Soda
- Department of Respiratory Medicine, Sasebo City General Hospital, Nagasaki, Japan
| | - Keisuke Iwasaki
- Department of Pathology, Sasebo City General Hospital, Nagasaki, Japan
| | - Takayuki Suyama
- Department of Respiratory Medicine, Sasebo City General Hospital, Nagasaki, Japan
| | - Hirokazu Taniguchi
- Department of Respiratory Medicine, Sasebo City General Hospital, Nagasaki, Japan.,Department of Respiratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Yuichi Fukuda
- Department of Respiratory Medicine, Sasebo City General Hospital, Nagasaki, Japan
| | - Hiroshi Mukae
- Department of Respiratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
195
|
Wang PF, Chen Y, Song SY, Wang TJ, Ji WJ, Li SW, Liu N, Yan CX. Immune-Related Adverse Events Associated with Anti-PD-1/PD-L1 Treatment for Malignancies: A Meta-Analysis. Front Pharmacol 2017; 8:730. [PMID: 29093678 PMCID: PMC5651530 DOI: 10.3389/fphar.2017.00730] [Citation(s) in RCA: 320] [Impact Index Per Article: 45.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 09/28/2017] [Indexed: 12/12/2022] Open
Abstract
Background: Treatment of cancers with programmed cell death protein 1 (PD-1) pathway inhibitors can lead to immune-related adverse events (irAEs), which could be serious and even fetal. Therefore, clinicians should be aware of the characteristics of irAEs associated with the use of such drugs. Methods: The MEDLINE, EMBASE, and Cochrane databases were searched to find potential studies using the following strategies: anti-PD-1/PD-L1 treatment; irAEs; and cancer. R© package Meta was used to pool incidence. Results: Forty-six studies representing 12,808 oncologic patients treated with anti-PD-1/PD-L1 agents were included in the meta-analysis. The anti-PD-1/PD-L1 agents included nivolumab, pembrolizumab, atezolizumab, durvalumab, avelumab, and BMS-936559. The tumor types were melanomas, Hodgkin lymphomas, urothelial carcinomas, breast cancers, non-small cell lung cancers, renal cell carcinomas (RCC), colorectal cancers, and others. We described irAEs according to organ systems, namely, the skin (pruritus, rash, maculopapular rash, vitiligo, and dermatitis), endocrine system (hypothyroidism, hyperthyroidism, hypophysitis, thyroiditis, and adrenal insufficiency), digestive system (colitis, diarrhea, pancreatitis, and increased AST/ALT/bilirubin), respiratory system (pneumonitis, lung infiltration, and interstitial lung disease), and urinary system (increased creatinine, nephritis, and renal failure). In patients treated with the PD-1 signaling inhibitors, the overall incidence of irAEs was 26.82% (95% CI, 21.73-32.61; I2, 92.80) in any grade and 6.10% (95% CI, 4.85-7.64; I2, 52.00) in severe grade, respectively. The development of irAEs was unrelated to the dose of anti-PD-1/PD-L1 agents. The incidence of particular irAEs varied when different cancers were treated with different drugs. The incidence of death due to irAEs was around 0.17%. Conclusion: The occurrence of irAEs was organ-specific and related to drug and tumor types.
Collapse
Affiliation(s)
- Peng-Fei Wang
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Yang Chen
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China.,State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Si-Ying Song
- Department of Clinical Medicine, Capital Medical University, Beijing, China
| | - Ting-Jian Wang
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Wen-Jun Ji
- Key Laboratory, Department of Neurosurgery, The Second Hospital of Yulin, Xi'an Jiaotong University, Xi'an, China
| | - Shou-Wei Li
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Ning Liu
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Chang-Xiang Yan
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
196
|
Hoshi S, Numahata K, Kanno H, Sato M, Kuromoto A, Nezu K, Sakai T, Konno C, Ishizuka Y, Izumi H, Taguchi K, Ono K, Hoshi K, Kanto S, Takahashi R, Vladimir B, Akimoto N, Sasagawa I, Ohta S. Updated recommendation on molecular-targeted therapy for metastatic renal cell cancer. Mol Clin Oncol 2017; 7:591-594. [PMID: 29046793 PMCID: PMC5639413 DOI: 10.3892/mco.2017.1371] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Accepted: 07/31/2017] [Indexed: 01/05/2023] Open
Abstract
Molecular-targeted therapy was recommended for the systemic therapy of renal cell cancer (RCC) in the RCC guidelines, but these guidelines do not address the order of administration of the multiple presently available agents. There are several aspects that remain unknown regarding the optimal administration order and combination of molecular-targeted drugs. Until the optimal treatment sequence is determined by clinical trials, treatment individualization is required for each patient based on patient and disease characteristics. We herein investigate 12 cases of RCC patients who received axitinib. Axitinib was used as the first-line drug in 4 cases, second-line in 5 cases, third-line in 1 case and as a fourth-line drug in 2 cases. Partial response (PR) was observed in 4 cases (30%) and stable disease in 4 cases (30%) during axitinib treatment, with an overall response rate of 60%. The duration of PR ranged from 6 to 19 months. Based on our cases, axitinib exhibited reasonable therapeutic efficacy as first- as well as second-line treatment. However, more cases are required to draw firm conclusions.
Collapse
Affiliation(s)
- Senji Hoshi
- Department of Urology, Yamagata Prefectural Central Hospital, Yamagata 990-2292, Japan
- Department Urology, Yamagata Tokushukai Hospital, Yamagata 990-0834, Japan
| | - Kenji Numahata
- Department of Urology, Yamagata Prefectural Central Hospital, Yamagata 990-2292, Japan
| | - Hidenori Kanno
- Department of Urology, Yamagata Prefectural Central Hospital, Yamagata 990-2292, Japan
| | - Masahiko Sato
- Department of Urology, Yamagata Prefectural Central Hospital, Yamagata 990-2292, Japan
| | - Akihito Kuromoto
- Department of Urology, Yamagata Prefectural Central Hospital, Yamagata 990-2292, Japan
| | - Kunihisa Nezu
- Department of Urology, Yamagata Prefectural Central Hospital, Yamagata 990-2292, Japan
| | - Takanari Sakai
- Department of Urology, Yamagata Prefectural Central Hospital, Yamagata 990-2292, Japan
| | - Chihito Konno
- Department of Urology, Ishinomaki Red Cross Hospital, Ishinomaki, Miyagi 986-8522, Japan
| | - Yuichi Ishizuka
- Department of Urology, Ishinomaki Red Cross Hospital, Ishinomaki, Miyagi 986-8522, Japan
| | - Hideaki Izumi
- Department of Urology, Ishinomaki Red Cross Hospital, Ishinomaki, Miyagi 986-8522, Japan
| | - Katsuyuki Taguchi
- Department of Urology, Ishinomaki Red Cross Hospital, Ishinomaki, Miyagi 986-8522, Japan
| | - Kunio Ono
- Department of Urology, Ishinomaki Red Cross Hospital, Ishinomaki, Miyagi 986-8522, Japan
| | - Kiyotsugu Hoshi
- Department Urology, Yamagata Tokushukai Hospital, Yamagata 990-0834, Japan
| | - Satoshi Kanto
- Department Urology, Yamagata Tokushukai Hospital, Yamagata 990-0834, Japan
| | - Rika Takahashi
- Department of Rehabilitation, Yamagata Tokushukai Hospital, Yamagata 990-0834, Japan
| | - Bilim Vladimir
- Department of Urology, Niigata Prefectural Cancer Center Hospital, Niigata 951-8566, Japan
| | - Naoe Akimoto
- Clinical Pathology, Faculty of Pharmaceutical Sciences, Josai University, Sakado, Saitama 350-0295, Japan
| | - Isoji Sasagawa
- Department Urology, Yamagata Tokushukai Hospital, Yamagata 990-0834, Japan
| | - Shoichiro Ohta
- Clinical Pathology, Faculty of Pharmaceutical Sciences, Josai University, Sakado, Saitama 350-0295, Japan
| |
Collapse
|
197
|
Brouwer NJ, Haanen JB, Jager MJ. Development of Ocular Rosacea following Combined Ipilimumab and Nivolumab Treatment for Metastatic Malignant Skin Melanoma. Ocul Oncol Pathol 2017; 3:188-192. [PMID: 29071268 PMCID: PMC5649334 DOI: 10.1159/000455150] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 12/01/2016] [Indexed: 12/16/2022] Open
Abstract
PURPOSE To report a case of severe ocular rosacea following ipilimumab plus nivolumab treatment in a patient with metastatic malignant skin melanoma. METHODS Case report and review of the literature. RESULTS A 68-year-old male with newly diagnosed metastatic malignant cutaneous melanoma was treated with first-line ipilimumab plus nivolumab, which resulted in a partial response. Four months after initiation of treatment, the patient developed red eyelids and conjunctivae, with painful gritty eyes, limiting his capacity to read. Following a diagnosis of severe ocular rosacea and dry eyes, treatment including corticosteroids, antimicrobial agents, and eyelid hygiene was started, and within 3 months, the ocular complaints resolved. CONCLUSION Treatment with checkpoint inhibitor immunotherapy for metastatic melanoma may trigger several ocular immune-related adverse events. This case describes severe ocular rosacea as an adverse event following ipilimumab plus nivolumab treatment.
Collapse
Affiliation(s)
- Niels J. Brouwer
- Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands
| | - John B.A.G. Haanen
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Martine J. Jager
- Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
198
|
Vanpouille-Box C, Lhuillier C, Bezu L, Aranda F, Yamazaki T, Kepp O, Fucikova J, Spisek R, Demaria S, Formenti SC, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: Immune checkpoint blockers for cancer therapy. Oncoimmunology 2017; 6:e1373237. [PMID: 29147629 DOI: 10.1080/2162402x.2017.1373237] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 08/25/2017] [Indexed: 02/08/2023] Open
Abstract
Immune checkpoint blockers (ICBs) are literally revolutionizing the clinical management of an ever more diversified panel of oncological indications. Although considerable attention persists around the inhibition of cytotoxic T lymphocyte-associated protein 4 (CTLA4) and programmed cell death 1 (PDCD1, best known as PD-1) signaling, several other co-inhibitory T-cell receptors are being evaluated as potential targets for the development of novel ICBs. Moreover, substantial efforts are being devoted to the identification of biomarkers that reliably predict the likelihood of each patient to obtain clinical benefits from ICBs in the absence of severe toxicity. Tailoring the delivery of specific ICBs or combinations thereof to selected patient populations in the context of precision medicine programs constitutes indeed a major objective of the future of ICB-based immunotherapy. Here, we discuss recent preclinical and clinical advances on the development of ICBs for oncological indications.
Collapse
Affiliation(s)
| | - Claire Lhuillier
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Lucillia Bezu
- Université Paris Descartes/Paris V, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,INSERM, U1138, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
| | - Fernando Aranda
- Immunoreceptors of the Innate and Adaptive System Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Takahiro Yamazaki
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Oliver Kepp
- Université Paris Descartes/Paris V, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,INSERM, U1138, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
| | - Jitka Fucikova
- Sotio a.c., Prague, Czech Republic.,Dept. of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czech Republic
| | - Radek Spisek
- Sotio a.c., Prague, Czech Republic.,Dept. of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czech Republic
| | - Sandra Demaria
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.,Sandra and Edward Meyer Cancer Center, New York, NY, USA
| | - Silvia C Formenti
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.,Sandra and Edward Meyer Cancer Center, New York, NY, USA
| | - Laurence Zitvogel
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,INSERM, U1015, Villejuif, France.,Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France.,Université Paris Sud/Paris XI, Le Kremlin-Bicêtre, France
| | - Guido Kroemer
- Université Paris Descartes/Paris V, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,INSERM, U1138, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,Department of Women's and Children's Health, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden.,Pôle de Biologie, Hopitâl Européen George Pompidou, AP-HP, Paris, France
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.,Université Paris Descartes/Paris V, Paris, France.,Sandra and Edward Meyer Cancer Center, New York, NY, USA
| |
Collapse
|
199
|
Ghatalia P, Zibelman M, Geynisman DM, Plimack ER. Checkpoint Inhibitors for the Treatment of Renal Cell Carcinoma. Curr Treat Options Oncol 2017; 18:7. [PMID: 28210995 DOI: 10.1007/s11864-017-0458-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OPINION STATEMENT The advent of checkpoint inhibitors has revolutionized systemic therapy for many malignancies, including renal cell carcinoma (RCC) where multiple PD-1, PD-L1, and CTLA-4 inhibitors have demonstrated responses and improved survival for patients in clinical trials. Durable benefit with manageable toxicity can be achieved with these agents-but unfortunately for only a minority of individuals. Efforts are ongoing to understand mechanisms driving the response and resistance to checkpoint inhibitors in order to personalize therapy and extend benefit to more patients. In particular, combination immunotherapy is an area of active study with multiple ongoing trials in RCC. Novel immunotherapeutic agents are being explored as well. Clinically, there are nuances related to the use of immunotherapy that are important to understand in order to provide optimal care to patients. Potential autoimmune toxicities are important to identify early so they can be best mitigated with immunosuppression, and careful review of imaging with clinical correlation is important to ensure responding patients are not taken off treatment prematurely due to "pseudo-progression." Lastly, although immunotherapy is an important new tool, it exists among other active agents in the treatment of RCC, and further study is needed to understand where it best fits in the treatment paradigm. In this article, we review the most recent data for immune checkpoint inhibitors in metastatic renal cell carcinoma and more broadly discuss the rapidly evolving landscape of immunotherapy in RCC, including combination immunotherapies.
Collapse
Affiliation(s)
- Pooja Ghatalia
- Department of Hematology/Oncology, Fox Chase Cancer Center, Temple Health, 333 Cottman Avenue, Philadelphia, PA, 19111-2497, USA
| | - Matthew Zibelman
- Department of Hematology/Oncology, Fox Chase Cancer Center, Temple Health, 333 Cottman Avenue, Philadelphia, PA, 19111-2497, USA
| | - Daniel M Geynisman
- Department of Hematology/Oncology, Fox Chase Cancer Center, Temple Health, 333 Cottman Avenue, Philadelphia, PA, 19111-2497, USA
| | - Elizabeth R Plimack
- Department of Hematology/Oncology, Fox Chase Cancer Center, Temple Health, 333 Cottman Avenue, Philadelphia, PA, 19111-2497, USA.
| |
Collapse
|
200
|
Gu L, Chen M, Guo D, Zhu H, Zhang W, Pan J, Zhong X, Li X, Qian H, Wang X. PD-L1 and gastric cancer prognosis: A systematic review and meta-analysis. PLoS One 2017; 12:e0182692. [PMID: 28796808 PMCID: PMC5552131 DOI: 10.1371/journal.pone.0182692] [Citation(s) in RCA: 183] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Accepted: 07/21/2017] [Indexed: 12/12/2022] Open
Abstract
The expression of Programmed cell Death Ligand 1 (PD-L1) is observed in many malignant tumors and is associated with poor prognosis including Gastric Cancer (GC). The relationship between PD-L1 expression and prognosis, however, is controversial in GC. This paper purports to use a meta-analysis to investigate the relationship between PD-L1 expression and prognosis in GC. For this study, the following databases were searched for articles published from June 2003 until February 2017: PubMed, EBSCO, Web of Science and Cochrane Library. The baseline information extracted were: authors, year of publication, country where the study was performed, study design, sample size, follow-up time, baseline characteristics of the study population, pathologic data, overall survival (OS). A total of 15 eligible studies covering 3291 patients were selected for a meta-analysis based on specified inclusion and exclusion criteria. The analysis showed that the expression level of PD-L1 was associated with the overall survival in GC (Hazard Ratio, HR = 1.46, 95%CI = 1.08-1.98, P = 0.01, random-effect). In addition to the above, subgroup analysis showed that GC patients with deeper tumor infiltration, positive lymph-node metastasis, positive venous invasion, Epstein-Barr virus infection positive (EBV+), Microsatellite Instability (MSI) are more likely to expression PD-L1. The results of this meta-analysis suggest that GC patients, specifically EBV+ and MSI, may be prime candidates for PD-1 directed therapy. These findings support anti-PD-L1/PD-1 antibodies as a kind of immunotherapy which is promising for GC.
Collapse
Affiliation(s)
- Lihu Gu
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Manman Chen
- The Second Clinical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Dongyu Guo
- Department of Ophthalmology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Hepan Zhu
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Wenchao Zhang
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Junhai Pan
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xin Zhong
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xinlong Li
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Haoran Qian
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xianfa Wang
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|