251
|
Redman JM, Rhea LP, Cordes L, Owens H, Madan RA, Bilusic M, Gulley JL, Lee JM, Dahut WL, Karzai F. A Case of Anti-PD-L1-associated Remitting Seronegative Symmetric Synovitis With Pitting Edema. Clin Genitourin Cancer 2019; 17:e549-e552. [PMID: 30850338 DOI: 10.1016/j.clgc.2019.02.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 02/05/2019] [Indexed: 12/17/2022]
Affiliation(s)
- Jason M Redman
- Medical Oncology Service, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Logan P Rhea
- Department of Medicine, Inova Fairfax Medical Campus, Falls Church, VA
| | - Lisa Cordes
- Genitourinary Malignancy Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Helen Owens
- Genitourinary Malignancy Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Ravi A Madan
- Genitourinary Malignancy Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Marijo Bilusic
- Genitourinary Malignancy Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - James L Gulley
- Genitourinary Malignancy Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Jung-Min Lee
- Women's Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - William L Dahut
- Genitourinary Malignancy Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Fatima Karzai
- Genitourinary Malignancy Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD.
| |
Collapse
|
252
|
Nakayama T, Kitano S. Immunotherapy for genitourinary tumors. Int J Urol 2019; 26:326-333. [PMID: 30710374 DOI: 10.1111/iju.13902] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 12/04/2018] [Indexed: 12/14/2022]
Abstract
The present review provides an update about the major achievements and recent advances of immunotherapy in renal cell carcinoma, urothelial carcinoma, and prostate cancer. Although the treatment strategy for renal cell carcinoma and urothelial carcinoma includes traditional cancer immunotherapies, such as interleukin-2 and interferon-alfa, the clinical outcomes of these therapies are unsatisfactory. In recent years, the development of immune checkpoint inhibitors has drastically changed the treatment strategy for various cancers, including genitourinary cancer. The present review summarizes the approved cancer immunotherapies for renal cell carcinoma, urothelial carcinoma and prostate cancer. Furthermore, we review the response evaluation and biomarkers for immune checkpoint inhibitors with a distinctive mode of action that is different from cytotoxic agents. Finally, future perspectives for cancer immunotherapy are discussed.
Collapse
Affiliation(s)
- Takayuki Nakayama
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
| | - Shigehisa Kitano
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
253
|
Ott PA, Bang YJ, Piha-Paul SA, Razak ARA, Bennouna J, Soria JC, Rugo HS, Cohen RB, O'Neil BH, Mehnert JM, Lopez J, Doi T, van Brummelen EMJ, Cristescu R, Yang P, Emancipator K, Stein K, Ayers M, Joe AK, Lunceford JK. T-Cell-Inflamed Gene-Expression Profile, Programmed Death Ligand 1 Expression, and Tumor Mutational Burden Predict Efficacy in Patients Treated With Pembrolizumab Across 20 Cancers: KEYNOTE-028. J Clin Oncol 2018; 37:318-327. [PMID: 30557521 DOI: 10.1200/jco.2018.78.2276] [Citation(s) in RCA: 648] [Impact Index Per Article: 92.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
PURPOSE Biomarkers that can predict response to anti-programmed cell death 1 (PD-1) therapy across multiple tumor types include a T-cell-inflamed gene-expression profile (GEP), programmed death ligand 1 (PD-L1) expression, and tumor mutational burden (TMB). Associations between these biomarkers and the clinical efficacy of pembrolizumab were evaluated in a clinical trial that encompassed 20 cohorts of patients with advanced solid tumors. METHODS KEYNOTE-028 ( ClinicalTrials.gov identifier: NCT02054806) is a nonrandomized, phase Ib trial that enrolled 475 patients with PD-L1-positive advanced solid tumors who were treated with pembrolizumab 10 mg/kg every 2 weeks for 2 years or until confirmed disease progression or unacceptable toxicity occurred. The primary end point was objective response rate (ORR; by RECIST v1.1, investigator review). Secondary end points included safety, progression-free survival (PFS), and overall survival (OS). Relationships between T-cell-inflamed GEP, PD-L1 expression, and TMB and antitumor activity were exploratory end points. RESULTS ORRs (with 95% CIs) ranged from 0% (0.0% to 14.2%) in pancreatic cancer to 33% (15.6% to 55.3%) in small-cell lung cancer. Across cohorts, median (95% CI) PFS ranged from 1.7 months (1.5 to 2.9 months) to 6.8 months (1.9 to 14.1 months) in pancreatic and thyroid cancers, respectively, and median OS from 3.9 months (2.8 to 5.5 months) to 21.1 months (9.1 to 22.4 months) in vulvar and carcinoid tumors, respectively. Higher response rates and longer PFS were demonstrated in tumors with higher T-cell-inflamed GEP, PD-L1 expression, and/or TMB. Correlations of TMB with GEP and PD-L1 were low. Response patterns indicate that patients with tumors that had high levels of both TMB and inflammatory markers (GEP or PD-L1) represent a population with the highest likelihood of response. Safety was similar and consistent with prior pembrolizumab reports. CONCLUSION A T-cell--inflamed GEP, PD-L1 expression, and TMB predicted response to pembrolizumab in multiple tumor types. These biomarkers (alone/in combination) may help identify patients who have a higher likelihood of response to anti-PD-1 therapies across a broad spectrum of cancers.
Collapse
Affiliation(s)
| | - Yung-Jue Bang
- Seoul National University College of Medicine, Seoul, The Republic of Korea
| | | | | | | | | | - Hope S Rugo
- University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, CA
| | | | | | | | - Juanita Lopez
- Institute of Cancer Research, London, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
254
|
Foster CC, Weichselbaum RR, Pitroda SP. Oligometastatic prostate cancer: Reality or figment of imagination? Cancer 2018; 125:340-352. [PMID: 30521067 PMCID: PMC6587458 DOI: 10.1002/cncr.31860] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 10/08/2018] [Accepted: 10/13/2018] [Indexed: 12/20/2022]
Abstract
The term “oligometastatic prostate cancer” refers to a heterogeneous group of disease states currently defined solely on the basis of clinical features. Oligorecurrent disease, de novo oligometastases, and oligoprogressive disease likely have unique biologic underpinnings and natural histories. Evidence suggesting the existence of a subset of patients who harbor prostate cancer with limited metastatic potential currently includes disparate and overwhelmingly retrospective reports. Nevertheless, emerging prospective data have corroborated the “better‐than‐expected,” retrospectively observed outcomes, particularly in the setting of oligorecurrent prostate cancer. Improved functional imaging with prostate‐specific membrane antigen‐targeted strategies may enhance the identification of patients with oligometastatic prostate cancer in the short term. In the long term, refinement of the oligometastatic case definition likely will require biologic risk‐stratification schemes. To determine optimal treatment strategies and identify patients most likely to benefit from metastasis‐directed therapy, future efforts should focus on conducting high‐quality, prospective trials with much‐needed molecular correlative studies. The term “oligometastatic prostate cancer” currently refers to a heterogeneous group of clinically defined disease states. To improve patient risk stratification and determine optimal treatment strategies, future efforts should focus on conducting prospective trials and determining a biologic categorization of patients with limited metastatic potential.
Collapse
Affiliation(s)
- Corey C Foster
- Department of Radiation and Cellular Oncology, The University of Chicago Medicine, Chicago, Illinois
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology, The University of Chicago Medicine, Chicago, Illinois
| | - Sean P Pitroda
- Department of Radiation and Cellular Oncology, The University of Chicago Medicine, Chicago, Illinois
| |
Collapse
|
255
|
Togasaki K, Sukawa Y, Kanai T, Takaishi H. Clinical efficacy of immune checkpoint inhibitors in the treatment of unresectable advanced or recurrent gastric cancer: an evidence-based review of therapies. Onco Targets Ther 2018; 11:8239-8250. [PMID: 30538493 PMCID: PMC6254591 DOI: 10.2147/ott.s152514] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Standard treatment options for patients with advanced gastric cancer (GC) offer limited efficacy and are associated with some toxicity, which necessitates the development of more effective therapies for improving the treatment outcomes for this disease. Immunotherapy involving immune checkpoint inhibitors (ICIs) which inhibit the programmed death 1 (PD-1)/programmed death ligand 1 interaction has emerged as a new treatment option. Nivolumab, a human IgG4 monoclonal antibody inhibitor of PD-1, has demonstrated promising clinical activity and induced durable responses in patients with advanced GC. Nivolumab has recently been approved for treating patients with pretreated advanced GC in Japan. In the present review, we summarized current evidence of the clinical efficacy of ICIs in a variety of solid tumors and reported our experience in patients with GC who were treated with nivolumab and the interesting features that were observed in these cases. Certain ICI-specific clinical features such as pseudo- and hyper-progression of tumor and hyper-response to subsequent chemotherapy have been reported in several cancer types. Lastly, we discussed the present scenario regarding research on biomarkers for assessing the clinical benefits of ICI therapies.
Collapse
Affiliation(s)
- Kazuhiro Togasaki
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan,
| | - Yasutaka Sukawa
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan,
| | - Takanori Kanai
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan,
| | - Hiromasa Takaishi
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan,
| |
Collapse
|
256
|
|
257
|
Rathke H, Flechsig P, Mier W, Bronzel M, Mavriopoulou E, Hohenfellner M, Giesel FL, Haberkorn U, Kratochwil C. Dosimetry Estimate and Initial Clinical Experience with 90Y-PSMA-617. J Nucl Med 2018; 60:806-811. [PMID: 30389816 DOI: 10.2967/jnumed.118.218917] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 10/22/2018] [Indexed: 01/17/2023] Open
Abstract
Because of different physical properties, the β-emitters 177Lu and 90Y offer specific radiologic-biologic advantages in dedicated clinical situations. Our objective was to introduce 90Y-labeled prostate-specific membrane antigen (PSMA)-617 to clinical application, providing additional avenues for personalized medicine. Here, we present our dosimetry estimate for 90Y-PSMA-617, report first clinical experiences, and discuss the advantages and drawbacks of varying the β-emitter in PSMA-targeting radioligand therapy. Methods: To approximate radiation dosimetry, 4 patients with metastatic castration-resistant prostate cancer underwent serially performed imaging up to 1 wk after 177Lu-PSMA-617 therapy. Time-activity curves were extrapolated to the half-life of 90Y, and OLINDA was used to calculate the dosimetry estimate. In clinical practice, 11 patients with PSMA-positive lymph-nodal bulk disease were stratified to receive 90Y-PSMA-617 radioligand therapy (mean, 3.2 GBq; range, 2.8-3.7 GBq); afterward, safety lab tests, prostate-specific antigen (PSA) response, and clinical findings were thoroughly followed. Results: The projected dosimetry for 90Y-PSMA-617 estimated a mean kidney dose of 3.47 ± 1.40 Gy/GBq, red marrow dose of 0.11 ± 0.04 Gy/GBq, and salivary gland dose of 5.57 ± 1.34 Gy/GBq; randomly chosen metastases were approximated with 22.8 ± 16.10 Gy/GBq. The observed acute hematologic toxicity (5 cases of leukopenia and 2 of thrombocytopenia, all grade 1 or 2) and clinical side effects (2 cases of transient xerostomia and 1 of nausea, all grade 1 or 2), as well as PSA response (any PSA response, 7/11 patients; >50% PSA decline, 5/11 patients), were comparable to 177Lu-PSMA-617 literature data. Conclusion: A factor 3-4 lower treatment activity for 90Y-PSMA-617 translates into a comparable dosimetry estimate and clinical findings similar to those of 177Lu-PSMA-617. However, safety was demonstrated only for patients with oligometastatic disease. Further studies are needed to evaluate its potential in patients with more disseminated bone involvement or visceral metastasis.
Collapse
Affiliation(s)
- Hendrik Rathke
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Paul Flechsig
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Walter Mier
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | | | - Eleni Mavriopoulou
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | | | - Frederik Lars Giesel
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Uwe Haberkorn
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany.,Cooperation Unit Nuclear Medicine, German Cancer Research Center, Heidelberg, Germany; and.,Translational Lung Research Center Heidelberg, German Center for Lung Research, Heidelberg, Germany
| | - Clemens Kratochwil
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
258
|
[Immunotherapy for the treatment of prostate cancer-a comeback?]. Urologe A 2018; 57:1342-1345. [PMID: 30341689 DOI: 10.1007/s00120-018-0790-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
BACKGROUND Prostate cancer (PCA) seems to be more of an immunologic desert than other tumor entities. It is striking that only rarely does prostate cancer show abundant immune cells and a proimmunogenic microenvironment. OBJECTIVES Is immunotherapy in PCA effective and which patients can benefit. MATERIALS AND METHODS A review of the literature and recent congress data are presented. RESULTS Preliminary results with sipuleucel-T for PCA cancer were very promising showing a significant overall survival benefit in randomised phase III studies and the US Federal Drug Administration (FDA) approval for this individualised vaccine. Contrary to other tumor entities this was not the immediate breakthrough to a new therapeutic era of immunotherapy but remained an isolated case and restricted to the USA. More recently, several trials evaluated immunotherapeutic agents but missed their preliminary endpoints. Interestingly, individual patients did benefit and showed long-term remission. CONCLUSIONS Genome sequencing and new biomarkers are also paving a novel pathway towards individualised immunotherapy for PCA. On-going research and clinical trials are exploring the question of which patients will benefit.
Collapse
|