51
|
Shi S, Wang H, Liu X, Xiao J. Prediction of overall survival of non-small cell lung cancer with bone metastasis: an analysis of the Surveillance, Epidemiology and End Results (SEER) database. Transl Cancer Res 2022; 10:5191-5203. [PMID: 35116369 PMCID: PMC8797363 DOI: 10.21037/tcr-21-1507] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 11/08/2021] [Indexed: 11/26/2022]
Abstract
Background The prognosis of non-small cell lung cancer (NSCLC) patients with bone metastasis is extremely repulsive. The aim of this study was to potentially characterize the prevalence, associated factors and to establish a prognostic nomogram to predict the overall survival (OS) of NSCLC patients with bone metastasis. Methods The Surveillance, Epidemiology and End Results (SEER) database was used to collected NSCLC cases during 2010–2015. The cases with incomplete clinicopathological information were excluded. Finally, 484 NSCLC patients with bone metastasis were included in the present study and randomly divided into the training (n=340) and validation (n=144) cohorts in a ratio of 7:3 based on R software. NSCLC patients with bone metastasis were selected to investigate predictive factors for OS and cancer-specific survival (CSS) using the multivariable Cox proportional hazards regression. A nomogram incorporating these prognostic factors was developed and evaluated by a concordance index (C-index), calibration plots, and risk group stratifications. Results In the Cox proportional hazards model, sex, race, American Joint Committee on Cancer (AJCC) N, T stage, liver metastasis, and chemotherapy were regarded as prognostic factors of OS. The nomogram based on sex, race, AJCC N, T stage, liver metastasis and chemotherapy was developed for cancer-specific death to predict 1-, 3-, and 5-year survival rate with good performance. The C-index of established nomogram was 0.695 for cancer-specific death in the study population with an acceptable calibration. Conclusions The female gender, the patients with chemotherapy and not liver metastasis may indicate improved survival. However, the global prospective data with the latest tumor, node, metastasis (TNM) classification is needed to further improve this model.
Collapse
Affiliation(s)
- Si Shi
- The Respiratory Department, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hongwei Wang
- The Respiratory Department, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiaohui Liu
- The Respiratory Department, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jinling Xiao
- The Respiratory Department, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
52
|
Huppert LA, Green MD, Kim L, Chow C, Leyfman Y, Daud AI, Lee JC. Tissue-specific Tregs in cancer metastasis: opportunities for precision immunotherapy. Cell Mol Immunol 2022; 19:33-45. [PMID: 34417572 PMCID: PMC8752797 DOI: 10.1038/s41423-021-00742-4] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 06/28/2021] [Indexed: 12/27/2022] Open
Abstract
Decades of advancements in immuno-oncology have enabled the development of current immunotherapies, which provide long-term treatment responses in certain metastatic cancer patients. However, cures remain infrequent, and most patients ultimately succumb to treatment-refractory metastatic disease. Recent insights suggest that tumors at certain organ sites exhibit distinctive response patterns to immunotherapy and can even reduce antitumor immunity within anatomically distant tumors, suggesting the activation of tissue-specific immune tolerogenic mechanisms in some cases of therapy resistance. Specialized immune cells known as regulatory T cells (Tregs) are present within all tissues in the body and coordinate the suppression of excessive immune activation to curb autoimmunity and maintain immune homeostasis. Despite the high volume of research on Tregs, the findings have failed to reconcile tissue-specific Treg functions in organs, such as tolerance, tissue repair, and regeneration, with their suppression of local and systemic tumor immunity in the context of immunotherapy resistance. To improve the understanding of how the tissue-specific functions of Tregs impact cancer immunotherapy, we review the specialized role of Tregs in clinically common and challenging organ sites of cancer metastasis, highlight research that describes Treg impacts on tissue-specific and systemic immune regulation in the context of immunotherapy, and summarize ongoing work reporting clinically feasible strategies that combine the specific targeting of Tregs with systemic cancer immunotherapy. Improved knowledge of Tregs in the framework of their tissue-specific biology and clinical sites of organ metastasis will enable more precise targeting of immunotherapy and have profound implications for treating patients with metastatic cancer.
Collapse
Affiliation(s)
- Laura A Huppert
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Michael D Green
- Department of Radiation Oncology, University of Michigan School of Medicine, Ann Arbor, MI, USA
- Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, MI, USA
| | - Luke Kim
- University of California, San Francisco School of Medicine, San Francisco, CA, USA
| | - Christine Chow
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Yan Leyfman
- Penn State College of Medicine, Hershey, PA, USA
| | - Adil I Daud
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - James C Lee
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA.
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA.
| |
Collapse
|
53
|
Jafari SH, Jahanmir A, Bahramvand Y, Tahmasebi S, Dallaki M, Nasrollahi E. Association of Estrogen Receptor, Progesterone Receptor, and Human Epidermal Growth Factor Receptor 2 Expression with Breast Cancer Metastasis in Iran. IRANIAN JOURNAL OF MEDICAL SCIENCES 2022; 47:40-47. [PMID: 35017776 PMCID: PMC8743368 DOI: 10.30476/ijms.2021.88366.1906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 01/27/2021] [Accepted: 02/16/2021] [Indexed: 11/23/2022]
Abstract
BACKGROUND Metastasis is an important factor in the survival estimate of patients with breast cancer. The present study aimed to examine the frequency of epidermal growth factor receptor 2 (HER2), estrogen receptor (ER), and progesterone receptor (PR) expression in relation to the metastatic site, pattern, and tumor size in patients with metastatic breast cancer (MBC). METHODS In this retrospective study, the medical records of patients diagnosed with MBC at Motahari Clinic (Shiraz, Iran) during 2017-2019 were examined. Metastasis was confirmed using computed tomography, and a total of 276 patients were included in the study. Based on the expression of receptors, the patients were categorized into luminal A, luminal B, HER2, and TNBC groups. The frequency and percentage of receptors in relation to the metastatic site, size, and pattern were compared using the Chi square test. P<0.05 was considered statistically significant. RESULTS The frequency of receptor positivity in the 276 selected medical records were of the subtype HER2-enriched (n=48), luminal A (n=43), luminal B (n=146), and TNBC (n=39). The most common metastatic sites were the bones (47.1%), lungs (34.4%), liver (27.9%), brain (20.3%), and other organs (12.7%). The first site of metastasis occurred in the bones (36.6%), lungs (17.4%), liver (15.6%), brain (10.5%), and other organs (7.6%). The frequency of receptor expression was different in relation to the first metastatic site (P=0.024). There was a statistically significant difference between the frequency of receptor expression in patients with bone (P=0.036), brain (P=0.031), and lung (P=0.020) metastases. The frequency of receptor expression was also significantly different in relation to the size of liver metastasis (P=0.009). Luminal A and B subtypes showed higher rates of bone metastasis as the first metastatic site. CONCLUSION The difference in the frequency of receptor expression in relation to the metastatic site and tumor size can be used as predictive and prognostic factors in patients with breast cancer.
Collapse
Affiliation(s)
- Seyed Hamed Jafari
- Medical Imaging Research Center, Shiraz University of Medical Sciences, Shiraz, Iran,
Department of Radiology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Armaghan Jahanmir
- Department of Radiology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Yaser Bahramvand
- Department of Radiology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sedigheh Tahmasebi
- Breast Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Manoochehr Dallaki
- Department of Radiology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Elham Nasrollahi
- Department of Radiology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
54
|
Safi M, Al-Azab M, Jin C, Trapani D, Baldi S, Adlat S, Wang A, Ahmad B, Al-madani H, Shan X, Liu J. Age-Based Disparities in Metastatic Melanoma Patients Treated in the Immune Checkpoint Inhibitors (ICI) Versus Non-ICI Era: A Population-Based Study. Front Immunol 2021; 12:609728. [PMID: 34887846 PMCID: PMC8650702 DOI: 10.3389/fimmu.2021.609728] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 09/20/2021] [Indexed: 12/11/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized metastatic melanoma treatment, but our knowledge of ICI activity across age groups is insufficient. Patients in different age groups with advanced melanoma were selected based on the ICI approval time in this study. Patients with melanoma were identified in the Surveillance, Epidemiology, and End Result (SEER) database program 2004-2016. The results showed that 4,040 patients had advanced melanoma before the advent of ICI (referred to as the "non-ICI era"), whereas there were 6,188 cases after ICI approval (referred to as the "ICI era"). In all age groups, the cases were dominated by men. The differences between the first (20-59 years) and second (60-74 years) age groups in both eras were significant in terms of surgery performance and holding of insurance policies (p = 0.05). The first and second groups (20-59 and 60-70 years old, respectively) showed no difference in survival (median = 8 months) during the non-ICI era, but the difference was evident in the first, second, and third age groups in the ICI era, with the younger group (20-59 years) having significantly better survival (median = 18, 14, and 10 months, respectively, p = 0.0001). Multivariate analysis of the first group (the youngest) in the ICI era revealed that surgery was significantly associated with an increase in survival among patients compared with those who did not undergo surgery (p < 0.0001). Furthermore, having an insurance policy among all age groups in the ICI era was associated with favorable survival in the first (20-59 years) and second (60-74 years) age groups (p = 0.0001), while there were no survival differences in the older ICI group (>74 years). Although there were differences in survival between the ICI era and the non-ICI era, these results demonstrate that ICI positively affected the survival of younger patients with advanced melanoma (first age group) than it had beneficial effects on older patients. Moreover, having had cancer surgery and holding an insurance policy were positive predictors for patient survival. This study emphasizes that adequate clinical and preclinical studies are important to enhance ICI outcomes across age groups.
Collapse
Affiliation(s)
- Mohammed Safi
- Department of Oncology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Mahmoud Al-Azab
- Department of Immunology, Guangzhou Women and Children’s Medical Centre, Guangzhou Medical University, Guangzhou, China
| | - Chenxing Jin
- Department of Oncology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | | | - Salem Baldi
- Department of Clinical Biochemistry, College of Laboratory Diagnostic Medicine, Dalian Medical University, Dalian, China
| | - Salah Adlat
- Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangzhou, China
| | - Aman Wang
- Department of Oncology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Bashir Ahmad
- Department of Biology, The University of Haripur, Haripur, Pakistan
| | - Hamza Al-madani
- Cixi Institute of Biomedical Engineering, Ningbo Institute of Materials Technology and Engineering, University of Chinese Academy of Sciences, Ningbo, China
| | - Xiu Shan
- Department of Oncology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jiwei Liu
- Department of Oncology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
55
|
Yin Q, Dai L, Sun R, Ke P, Liu L, Jiang B. Clinical Efficacy of Immune Checkpoint Inhibitors in Non-small-cell Lung Cancer Patients with Liver Metastases: A Network Meta-Analysis of Nine Randomized Controlled Trials. Cancer Res Treat 2021; 54:803-816. [PMID: 34696564 PMCID: PMC9296924 DOI: 10.4143/crt.2021.764] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 10/21/2021] [Indexed: 11/21/2022] Open
Abstract
Purpose This network meta-analysis (NMA) was conducted to compare the efficacy of immune checkpoint inhibitors (ICIs) in advanced non-small cell lung cancer (NSCLC) patients with liver metastases. Materials and Methods English literature was retrieved from the PubMed, American Society of Clinical Oncology (ASCO), and European Society for Medical Oncology (ESMO) databases from January 2015 to January 2021. We pooled the overall survival (OS) and progression-free survival (PFS) hazard ratios (HRs) using a network meta-analysis and ranked treatments by the surface under the cumulative ranking curve (SUCRA). Publication bias was evaluated by Begg's and Egger's tests. STATA15.0 was used for the sensitivity analysis, and the remaining statistical analyses were performed using R 4.0.2. Results Nine eligible phase III randomized controlled trials (RCTs) were included, including 1,141 patients with liver metastases. Pembrolizumab + chemotherapy ranked highest, followed by atezolizumab + bevacizumab + chemotherapy and nivolumab. However, no significant difference in OS rates was observed across these three treatments (HR, 0.98; 95% CI: 0.43-2.22 for pembrolizumab + chemotherapy vs. atezolizumab + bevacizumab + chemotherapy; HR, 0.91; 95% CI: 0.52-1.57 for pembrolizumab + chemotherapy vs. nivolumab). Regarding the PFS rate, atezolizumab + bevacizumab + chemotherapy and pembrolizumab + chemotherapy ranked highest and no significant difference was observed between them (HR, 0.79; 95% CI: 0.36-1.70 for atezolizumab + bevacizumab + chemotherapy vs. pembrolizumab + chemotherapy). Conclusion Pembrolizumab + chemotherapy, atezolizumab + bevacizumab + chemotherapy, and nivolumab were superior to other treatments in NSCLC patients with liver metastases. These new findings may help clinicians better select therapeutic strategies for NSCLC patients with liver metastases.
Collapse
Affiliation(s)
- Qing Yin
- Cadre Medical Department, The Third Affiliated Hospital of Kunming Medical University, Yunnan, China
| | - Longguo Dai
- Cadre Medical Department, The Third Affiliated Hospital of Kunming Medical University, Yunnan, China
| | - Ruizhu Sun
- Cadre Medical Department, The Third Affiliated Hospital of Kunming Medical University, Yunnan, China
| | - Ping Ke
- Cadre Medical Department, The Third Affiliated Hospital of Kunming Medical University, Yunnan, China
| | - Liya Liu
- Cadre Medical Department, The Third Affiliated Hospital of Kunming Medical University, Yunnan, China
| | - Bo Jiang
- Cadre Medical Department, The Third Affiliated Hospital of Kunming Medical University, Yunnan, China
| |
Collapse
|
56
|
Kim CW, Chon HJ, Kim C. Combination Immunotherapies to Overcome Intrinsic Resistance to Checkpoint Blockade in Microsatellite Stable Colorectal Cancer. Cancers (Basel) 2021; 13:4906. [PMID: 34638390 PMCID: PMC8507875 DOI: 10.3390/cancers13194906] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 09/18/2021] [Accepted: 09/26/2021] [Indexed: 12/19/2022] Open
Abstract
Although immune checkpoint inhibitors (ICIs) have shown promising results in the treatment of treating various malignancies, progress has been severely limited in metastatic colorectal cancer (mCRC). ICIs are effective in a fraction of patients with microsatellite instability-high mCRC but have little clinical efficacy in patients with microsatellite stable (MSS) mCRC, which accounts for 95% of mCRC cases. MSS mCRCs are considered to have intrinsic resistance to ICI monotherapy through multiple mechanisms. (1) They are poorly immunogenic because of their low tumor mutation burden; (2) frequent activation of the WNT/β-catenin signaling pathway excludes intratumoral CD8+ T cell immunity; (3) the tumor microenvironment is immunosuppressive because of the presence of various immunosuppressive cells, including tumor-associated macrophages and regulatory T cells; and (4) frequent liver metastasis in MSS mCRC may reduce the efficacy of ICIs. To overcome these resistance mechanisms, combination approaches using various agents, including STING agonists, MEK inhibitors, VEGF/R inhibitors, WNT/β-catenin inhibitors, oncolytic viruses, and chemo/radiotherapy, are actively ongoing. Preliminary evidence of the efficacy of some has been shown in early clinical trials. This review summarizes novel combination immunotherapy strategies described in recent preclinical and clinical studies to overcome the limitations of ICI monotherapy in MSS mCRC.
Collapse
Affiliation(s)
- Chang Woo Kim
- Department of Surgery, Ajou University School of Medicine, 164 World Cup-ro, Yeongtong-gu, Suwon 16499, Korea;
| | - Hong Jae Chon
- Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, 59 Yatap-ro, Bundang-gu, Seongnam 13496, Korea
| | - Chan Kim
- Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, 59 Yatap-ro, Bundang-gu, Seongnam 13496, Korea
| |
Collapse
|
57
|
Affiliation(s)
- Chandan Guha
- Departments of Radiation Oncology, Pathology and Urology, and Institute of Onco-Physics, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY.
| |
Collapse
|
58
|
Botticelli A, Cirillo A, Strigari L, Valentini F, Cerbelli B, Scagnoli S, Cerbelli E, Zizzari IG, Rocca CD, D’Amati G, Polimeni A, Nuti M, Merlano MC, Mezi S, Marchetti P. Anti-PD-1 and Anti-PD-L1 in Head and Neck Cancer: A Network Meta-Analysis. Front Immunol 2021; 12:705096. [PMID: 34434192 PMCID: PMC8380817 DOI: 10.3389/fimmu.2021.705096] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 07/12/2021] [Indexed: 12/21/2022] Open
Abstract
Objective The monoclonal antibodies anti-programmed death protein-1 (anti-PD-1) nivolumab and pembrolizumab are the first immune checkpoint inhibitors (ICIs) approved for treatment of recurrent/metastatic head and neck carcinoma R/M HNSCC in first line and in platinum refractory disease. This network meta-analysis aims to investigate the efficacy of anti-PD-1- vs anti-PD-L1-based therapy in R/M HNSCC cancer patients through a systematic review of the literature to provide support for evidence-based treatment decisions. In particular, the effectiveness of ICIs for R/M HNSCC is analyzed according to the different mechanisms of action of the check-points inhibitory drugs in different subgroups of patients. Methods We did a systematic literature review and network meta-analysis (NMA) of randomized controlled trials (RCTs) in PubMed, ClinicalTrials.gov, Embase, Medline, the Cochrane Central Register of Controlled Trials, Web of Science. Our search identified a total of five randomized controlled trials: Keynote 040, Keynote 048, Eagle, Condor, Checkmate 141. These trials included 3001 patients. Treatment was sub-categorized into PD-L1-based, PD-1-based, and standard chemotherapy. Treatments were indirectly compared with anti-PD-L1-based therapy. Results The network meta-analysis demonstrated no significant differences in OS between different subgroups except for the metastatic patients in which anti-PD-1-based therapy was associated with significantly less risk of death. Furthermore, anti-PD-1-based therapy appeared to be effective in smoker patients and in human papilloma-negative (HPV) patients. Conversely, anti-PD-L1-based therapy seems to be better efficient in female patients, in locally recurrent setting and in HPV positive patients. Conclusion This is the first NMA study that aimed to indirectly compare anti-PD-1- and anti-PD-L1-based therapy in HNSCC patients. The results of our NMA could help define a profile of patient responder or resistant to specific classes of immune drugs and can be used to guide/design future studies in the novel scenario of precision immune-oncology.
Collapse
Affiliation(s)
- Andrea Botticelli
- Department of Clinical and Molecular Oncology, “Sapienza” University of Rome, Rome, Italy
| | - Alessio Cirillo
- Department of Radiological, Oncological, and Anatomo-Pathological Science “Sapienza”, University of Rome, Rome, Italy
| | - Lidia Strigari
- Medical Physics Unit, “S. Orsola-Malpighi” Hospital, Bologna, Italy
| | - Filippo Valentini
- Department of Radiological, Oncological, and Anatomo-Pathological Science “Sapienza”, University of Rome, Rome, Italy
| | - Bruna Cerbelli
- Department of Radiological, Oncological, and Anatomo-Pathological Science “Sapienza”, University of Rome, Rome, Italy
| | - Simone Scagnoli
- Department of Radiological, Oncological, and Anatomo-Pathological Science “Sapienza”, University of Rome, Rome, Italy
| | - Edoardo Cerbelli
- Department of Radiological, Oncological, and Anatomo-Pathological Science “Sapienza”, University of Rome, Rome, Italy
| | | | - Carlo Della Rocca
- Department of Medico-Surgical Sciences and Biotechnology, Polo Pontino, Sapienza University, Roma, Italy
| | - Giulia D’Amati
- Department of Radiological, Oncological, and Anatomo-Pathological Science “Sapienza”, University of Rome, Rome, Italy
| | - Antonella Polimeni
- Odontostomatological and Maxillo-Facial Science, ‘Sapienza’ University of Rome, Rome, Italy
| | - Marianna Nuti
- Department of Experimental Medicine, University Sapienza, Rome, Italy
| | - Marco Carlo Merlano
- Medical Oncology, Candiolo Cancer Institute, FPO-IRCCS, Candiolo (Turin), Italy
| | - Silvia Mezi
- Department of Radiological, Oncological, and Anatomo-Pathological Science “Sapienza”, University of Rome, Rome, Italy
| | - Paolo Marchetti
- Department of Clinical and Molecular Oncology, “Sapienza” University of Rome, Rome, Italy
| |
Collapse
|
59
|
Borgers JSW, Tobin RP, Torphy RJ, Vorwald VM, Van Gulick RJ, Amato CM, Cogswell DT, Chimed TS, Couts KL, Van Bokhoven A, Raeburn CD, Lewis KD, Wisell J, McCarter MD, Mushtaq RR, Robinson WA. Melanoma Metastases to the Adrenal Gland Are Highly Resistant to Immune Checkpoint Inhibitors. J Natl Compr Canc Netw 2021; 19:jnccn20283. [PMID: 34348236 DOI: 10.6004/jnccn.2020.7800] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 12/16/2020] [Indexed: 12/07/2022]
Abstract
BACKGROUND Adrenal gland metastases (AGMs) are common in advanced-stage melanoma, occurring in up to 50% of patients. The introduction of immune checkpoint inhibitors (ICIs) has markedly altered the outcome of patients with melanoma. However, despite significant successes, anecdotal evidence has suggested that treatment responses in AGMs are significantly lower than in other metastatic sites. We sought to investigate whether having an AGM is associated with altered outcomes and whether ICI responses are dampened in the adrenal glands. PATIENTS AND METHODS We retrospectively compared ICI responses and overall survival (OS) in 68 patients with melanoma who were diagnosed with an AGM and a control group of 100 patients without AGMs at a single institution. Response was determined using RECIST 1.1. OS was calculated from time of ICI initiation, anti-PD-1 initiation, initial melanoma diagnosis, and stage IV disease diagnosis. Tumor-infiltrating immune cells were characterized in 9 resected AGMs using immunohistochemical analysis. RESULTS Response rates of AGMs were significantly lower compared with other metastatic sites in patients with AGMs (16% vs 22%) and compared with those without AGMs (55%). Patients with AGMs also had significantly lower median OS compared with those without AGMs (3.1 years vs not reached, respectively). We further observed that despite this, AGMs exhibited high levels of tumor-infiltrating immune cells. CONCLUSIONS In this cohort of patients with melanoma, those diagnosed with an AGM had lower ICI response rates and OS. These results suggest that tissue-specific microenvironments of AGMs present unique challenges that may require novel, adrenal gland-directed therapies or surgical resection.
Collapse
Affiliation(s)
- Jessica S W Borgers
- 1Division of Surgical Oncology, Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- 2The Netherlands Cancer Institute, Amsterdam, the Netherlands; and
| | - Richard P Tobin
- 1Division of Surgical Oncology, Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- 3Center for Rare Melanomas
- 4International Melanoma Biorepository, Center for Rare Melanomas
| | - Robert J Torphy
- 1Division of Surgical Oncology, Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Victoria M Vorwald
- 1Division of Surgical Oncology, Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- 3Center for Rare Melanomas
- 4International Melanoma Biorepository, Center for Rare Melanomas
| | - Robert J Van Gulick
- 3Center for Rare Melanomas
- 4International Melanoma Biorepository, Center for Rare Melanomas
- 5Division of Medical Oncology, Department of Medicine
| | - Carol M Amato
- 3Center for Rare Melanomas
- 4International Melanoma Biorepository, Center for Rare Melanomas
- 5Division of Medical Oncology, Department of Medicine
| | - Dasha T Cogswell
- 1Division of Surgical Oncology, Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- 3Center for Rare Melanomas
- 4International Melanoma Biorepository, Center for Rare Melanomas
| | | | - Kasey L Couts
- 4International Melanoma Biorepository, Center for Rare Melanomas
- 5Division of Medical Oncology, Department of Medicine
| | | | - Christopher D Raeburn
- 7Division of GI, Trauma, and Endocrine Surgery, Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Karl D Lewis
- 4International Melanoma Biorepository, Center for Rare Melanomas
- 5Division of Medical Oncology, Department of Medicine
| | - Joshua Wisell
- 4International Melanoma Biorepository, Center for Rare Melanomas
- 6Department of Pathology, and
| | - Martin D McCarter
- 1Division of Surgical Oncology, Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- 3Center for Rare Melanomas
- 4International Melanoma Biorepository, Center for Rare Melanomas
| | - Rao R Mushtaq
- 5Division of Medical Oncology, Department of Medicine
| | - William A Robinson
- 3Center for Rare Melanomas
- 4International Melanoma Biorepository, Center for Rare Melanomas
- 5Division of Medical Oncology, Department of Medicine
| |
Collapse
|
60
|
Wang C, Sandhu J, Ouyang C, Ye J, Lee PP, Fakih M. Clinical Response to Immunotherapy Targeting Programmed Cell Death Receptor 1/Programmed Cell Death Ligand 1 in Patients With Treatment-Resistant Microsatellite Stable Colorectal Cancer With and Without Liver Metastases. JAMA Netw Open 2021; 4:e2118416. [PMID: 34369992 PMCID: PMC8353537 DOI: 10.1001/jamanetworkopen.2021.18416] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
IMPORTANCE Microsatellite stable (MSS) metastatic colorectal cancer has been historically characterized as resistant to immunotherapy. Recent studies have demonstrated limited clinical activity of programmed cell death receptor 1/programmed death ligand 1 (PD-1/PD-L1) targeting in MSS metastatic colorectal cancer. The association of metastatic disease in the liver with treatment response has not been fully investigated. OBJECTIVE To investigate the association of liver metastases with response to PD-1/PD-L1-targeting therapy in MSS metastatic colorectal cancer. DESIGN, SETTING, AND PARTICIPANTS This single-center retrospective cohort study evaluated clinical responses to PD-1- or PD-L1-targeting therapy, with or without other investigational agents, in patients with MSS metastatic colorectal cancer and disease progression after standard of care therapy from January 1, 2014, to December 31, 2020. MAIN OUTCOMES AND MEASURES Objective response rate (ORR) and progression-free survival (PFS), measured from initiation of PD-1/PD-L1-targeting therapy. RESULTS Ninety-five patients with MSS metastatic colorectal cancer were identified (54 men [56.8%]; median age, 55 [interquartile range (IQR), 49-64] years). The overall ORR was 8.4% (8 of 95 patients). Eight of 41 patients without liver metastases achieved an ORR of 19.5%, and no response was observed in 54 patients with liver metastases. The disease control rate was 58.5% (24 of 41) in patients without liver metastasis and 1.9% (1 of 54) in patients with liver metastasis. Patients without liver metastases at the time of PD-1/PD-L1-targeting treatment had a superior median PFS compared with patients with liver metastases (4.0 [IQR, 2.0-7.5] vs 1.5 [IQR, 1.0-2.0] months; P < .001). In addition, median PFS was 5.5 (IQR, 2.0-11.5) months for patients without any prior or current liver involvement at the time of PD-1/PD-L1-targeting treatment initiation. Using a multivariate Cox regression model correcting for Eastern Cooperative Oncology Group status, primary tumor location, RAS and BRAF status, tumor mutation burden, and metastatic sites, liver metastases was the variable with the most significant association with faster progression after PD-1/PD-L1 treatment inhibition (hazard ratio, 7.00; 95% CI, 3.18-15.42; P < .001). CONCLUSIONS AND RELEVANCE Findings of this cohort study suggest that patients with MSS metastatic colorectal cancer and without liver metastases may derive clinical benefits from checkpoint inhibitors, whereas the presence of liver metastases was associated with resistance. Further prospective studies are needed to investigate PD-1/PD-L1 inhibitors in patients with MSS metastatic colorectal cancer without liver metastases.
Collapse
Affiliation(s)
- Chongkai Wang
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Jaideep Sandhu
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Ching Ouyang
- Center for Informatics, City of Hope National Medical Center, Duarte, California
- Department of Computational and Quantitative Medicine, Beckman Research Institute of the City of Hope, Duarte, California
| | - Jian Ye
- Department of Immuno-oncology, Beckman Research Institute of the City of Hope, Duarte, California
| | - Peter P. Lee
- Department of Immuno-oncology, Beckman Research Institute of the City of Hope, Duarte, California
| | - Marwan Fakih
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, California
| |
Collapse
|
61
|
Miller KM, Filippova OT, Hayes SA, Abu-Rustum NR, Aghajanian C, Broach V, Ellenson LH, Selenica P, Jewell EL, Kyi C, Lakhman Y, Mueller JJ, O'Cearbhaill RE, Park KJ, Sonoda Y, Zamarin D, Weigelt B, Leitao MM, Friedman CF. Pattern of disease and response to pembrolizumab in recurrent cervical cancer. Gynecol Oncol Rep 2021; 37:100831. [PMID: 34345644 PMCID: PMC8319446 DOI: 10.1016/j.gore.2021.100831] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 06/28/2021] [Accepted: 07/06/2021] [Indexed: 12/20/2022] Open
Abstract
Objective Since the approval of pembrolizumab for advanced or recurrent PD-L1 positive (CPS > 1%) cervical cancer, the clinical characteristics associated with response have remained undefined. We sought to characterize the clinicopathologic features of patients with advanced cervical cancer at our institution who derived durable clinical benefit from treatment with pembrolizumab. Methods We conducted a retrospective cohort study of 14 patients with recurrent or metastatic cervical cancer who received pembrolizumab monotherapy from August 2017 to November 2019 and were followed until November 1, 2020. Reviewed clinical data included age, histology, tumor molecular profiling results, stage at diagnosis, treatment history, baseline pattern of metastatic disease at initiation of anti-PD-1 therapy, and outcomes. Treatment response was evaluated by computed tomography using RECIST v1.1 criteria. Results The objective response rate was 21% (n = 3), including two partial responses and one complete response. Two patients (14%) had stable disease of six months or greater, for an observed durable clinical benefit rate of 36%. When stratified by those who derived clinical benefit, metastatic spread to lung and/or lymph node only at baseline was associated with improved response to pembrolizumab (n = 7, p = 0.02) and associated with significantly improved PFS and OS. Tumor mutational burden was higher in those with durable clinical benefit compared to non-responders (median 12.7 vs. 3.5 mutations/megabase, p = 0.03). Conclusions Our findings highlight clinical features that may select for a population most likely to benefit from pembrolizumab monotherapy and underscores the need for identification of additional biomarkers of response.
Collapse
Affiliation(s)
- Kathryn M Miller
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Olga T Filippova
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Sara A Hayes
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Nadeem R Abu-Rustum
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States.,Department of Surgery, Weill Cornell Medical College, New York, NY, United States
| | - Carol Aghajanian
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States.,Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| | - Vance Broach
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States.,Department of Surgery, Weill Cornell Medical College, New York, NY, United States
| | - Lora H Ellenson
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Pier Selenica
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Elizabeth L Jewell
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States.,Department of Surgery, Weill Cornell Medical College, New York, NY, United States
| | - Chrisann Kyi
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States.,Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| | - Yuliya Lakhman
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Jennifer J Mueller
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States.,Department of Surgery, Weill Cornell Medical College, New York, NY, United States
| | - Roisin E O'Cearbhaill
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States.,Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| | - Kay J Park
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Yukio Sonoda
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States.,Department of Surgery, Weill Cornell Medical College, New York, NY, United States
| | - Dmitriy Zamarin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States.,Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| | - Britta Weigelt
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Mario M Leitao
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States.,Department of Surgery, Weill Cornell Medical College, New York, NY, United States
| | - Claire F Friedman
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States.,Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| |
Collapse
|
62
|
Hwang I, Park I, Yoon SK, Lee JL. Clinical course of patients with renal cell carcinoma or urothelial carcinoma who had stable disease as an initial response to a PD-1 or PD-L1 inhibitor. Asia Pac J Clin Oncol 2021; 18:371-377. [PMID: 34219393 DOI: 10.1111/ajco.13601] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 04/06/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND About 17%-34% of patients with non-prostatic genitourinary (GU) cancer had stable disease (SD) as their best response to programmed cell death protein 1/programmed death-ligand 1 (PD-1/PD-L1) inhibitors. However, most efficacy studies of PD-1/PD-L1 inhibitors are focused on the response and its durability. Little is known about patients' clinical outcomes with SD as their initial response to PD-1/PD-L1 inhibitors. METHODS We retrospectively reviewed the clinical outcomes of patients with renal cell carcinoma (RCC) (n = 102) and urothelial carcinoma (UC) (n = 101) treated with PD-1/PD-L1 inhibitors. The duration of SD (DoSD) was calculated for patients who had SD as their best response and was defined from the time of SD to radiologically confirmed progressive disease (PD) or death. RESULTS Fifty-five patients (27.1%) had SD as the initial response. Among them, 10 patients (18.2%) achieved a response on subsequent assessments. With a median follow-up duration of 15.2 months, the median DoSD was 8.5 months (range: 3.2-13.8), which was significantly different according to the disease; 9.4 months in RCC and 2.3 months in UC (p = 0.03). If the disease did not progress in 4 months (long SD subgroup), the DoSD (11.0 months) was comparable to the duration of response (12.9 months) in patients who achieved a complete or partial response to PD-1/PD-L1 inhibitors. In addition to the cancer type, the clinical factors associated with short DoSD were liver metastases (p = 0.003), neutrophil-lymphocyte ratio (NLR) > 4.0 (p = 0.001), and platelet-lymphocyte ratio (PLR) > 194 (p = 0.003). CONCLUSION For RCC patients with SD to PD-1/PD-L1 inhibitors, if there are no liver metastases and NLR/PLR is below the certain level, they would have similar duration of disease control and survival benefit as those who achieve the response.
Collapse
Affiliation(s)
- Inhwan Hwang
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.,Department of Oncology, Yuseong Sun Medical Center, Daejeon, South Korea
| | - Inkeun Park
- Division of Medical Oncology, Department of Internal Medicine, Gil Medical Center, Gachon University School of Medicine, Incheon, Korea
| | - Shin-Kyo Yoon
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jae Lyun Lee
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
63
|
Wang C, Fakih M. Targeting MSS colorectal cancer with immunotherapy: are we turning the corner? Expert Opin Biol Ther 2021; 21:1347-1357. [PMID: 34030532 DOI: 10.1080/14712598.2021.1933940] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Immunotherapy with checkpoint inhibition has shown potent antitumor activity in patients with microsatellite instability (MSI) metastatic cancer. Microsatellite stable (MSS) colorectal cancer has long been considered resistant to immunotherapy. AREAS COVERED In this review, we provide an overview of current progress on strategies to overcome the resistance to immunotherapy in MSS colorectal cancer. EXPERT OPINION Emerging evidence suggest that combination of immune modulators such as regorafenib may improve the responsiveness of MSS colorectal cancer to checkpoint blockade. In addition, signs of clinical activity have also been observed in other combination strategies, such as the combination of checkpoint blockade with Stat3 inhibitor, or bispecific T-cell engagers. Nevertheless, predictive biomarkers that can identify patients who may benefit from immunotherapy are key for its implementation in clinical setting. Metastatic disease sites may predict for the response or resistance to checkpoint blockade, with liver metastases emerging as a strong predictive biomarker of lack of benefit from PD-1 targeting, even with combination therapies. Additional efforts are required to study the mechanism of resistance and to develop novel therapeutic strategies to overcome immune resistance. ABBREVIATIONS CEA: carcinoembryonic antigen; CR: complete response; CTLA-4: cytotoxic T-lymphocyte-associated protein 4; DCR: disease control rate; MSI-H: microsatellite instability-high; MSS: Microsatellite stable (MSS); OS: overall survival; PD-1: programmed cell death protein 1; PD-L1: programmed death-ligand receptor 1; PR: partial response; PFS: progression-free survival; SD: stable disease; TMB: tumor mutation burden; VEGFR: vascular endothelial growth factor receptor.
Collapse
Affiliation(s)
- Chongkai Wang
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Marwan Fakih
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| |
Collapse
|
64
|
Peng L, Zhang Y, Wang Z. Immune Responses against Disseminated Tumor Cells. Cancers (Basel) 2021; 13:2515. [PMID: 34063848 PMCID: PMC8196619 DOI: 10.3390/cancers13112515] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/17/2021] [Accepted: 05/19/2021] [Indexed: 02/07/2023] Open
Abstract
Most cancer-related deaths are a consequence of metastases, a series of linear events, notably the invasion-metastasis cascade. The current understanding of cancer immune surveillance derives from studies in primary tumors, but disseminated cancer cells acquire mutations and, in some cases, appear to progress independently after spreading from primary sites. An early step in this process is micrometastatic dissemination. As such, the equilibrium between the immune system and disseminated cancer cells controls the fate of the cancer. Immune checkpoint inhibitors (ICIs) exhibit significant clinical activity in patients, but the efficacy of ICIs depends on both the tumor and its microenvironment. Data often suggest that disseminated cancer cells are not adequately targeted by the immune system. In this review, we summarize the main basic findings of immune responses against disseminated tumor cells and their organ-specific characteristics. Such studies may provide new directions for cancer immune therapy.
Collapse
Affiliation(s)
- Ling Peng
- Department of Pulmonary and Critical Care Medicine, Zhejiang Provincial People’s Hospital, Hangzhou 310014, China
| | - Yongchang Zhang
- Lung Cancer and Gastrointestinal Unit, Department of Medical Oncology, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Changsha 410013, China;
| | - Zibing Wang
- Department of Immunotherapy, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China
| |
Collapse
|
65
|
Tang Y, Li X, Cao Y. Which factors matter the most? Revisiting and dissecting antibody therapeutic doses. Drug Discov Today 2021; 26:1980-1990. [PMID: 33895315 DOI: 10.1016/j.drudis.2021.04.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 02/28/2021] [Accepted: 04/16/2021] [Indexed: 01/22/2023]
Abstract
Factors such as antibody clearance and target affinity can influence antibodies' effective doses for specific indications. However, these factors vary considerably across antibody classes, precluding direct and quantitative comparisons. Here, we apply a dimensionless metric, the therapeutic exposure affinity ratio (TEAR), which normalizes the therapeutic doses by antibody bioavailability, systemic clearance and target-binding property to enable direct and quantitative comparisons of therapeutic doses. Using TEAR, we revisited and dissected the doses of up to 60 approved antibodies. We failed to detect a significant influence of target baselines, turnovers or anatomical locations on antibody therapeutic doses, challenging the traditional perceptions. We highlight the importance of antibodies' modes of action for therapeutic doses and dose selections; antibodies that work through neutralizing soluble targets show higher TEARs than those working through other mechanisms. Overall, our analysis provides insights into the factors that influence antibody doses, and the factors that are crucial for antibodies' pharmacological effects.
Collapse
Affiliation(s)
- Yu Tang
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Xiaobing Li
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yanguang Cao
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
66
|
Abstract
Liver metastases are commonly detected in a range of malignancies including colorectal cancer (CRC), pancreatic cancer, melanoma, lung cancer and breast cancer, although CRC is the most common primary cancer that metastasizes to the liver. Interactions between tumour cells and the tumour microenvironment play an important part in the engraftment, survival and progression of the metastases. Various cells including liver sinusoidal endothelial cells, Kupffer cells, hepatic stellate cells, parenchymal hepatocytes, dendritic cells, resident natural killer cells as well as other immune cells such as monocytes, macrophages and neutrophils are implicated in promoting and sustaining metastases in the liver. Four key phases (microvascular, pre-angiogenic, angiogenic and growth phases) have been identified in the process of liver metastasis. Imaging modalities such as ultrasonography, CT, MRI and PET scans are typically used for the diagnosis of liver metastases. Surgical resection remains the main potentially curative treatment among patients with resectable liver metastases. The role of liver transplantation in the management of liver metastasis remains controversial. Systemic therapies, newer biologic agents (for example, bevacizumab and cetuximab) and immunotherapeutic agents have revolutionized the treatment options for liver metastases. Moving forward, incorporation of genetic tests can provide more accurate information to guide clinical decision-making and predict prognosis among patients with liver metastases.
Collapse
|
67
|
Do OA, Ferris LA, Holt SK, Ramos JD, Harshman LC, Plimack ER, Crabb SJ, Pal SK, De Giorgi U, Ladoire S, Baniel J, Necchi A, Vaishampayan UN, Bamias A, Bellmunt J, Srinivas S, Dorff TB, Galsky MD, Yu EY. Treatment of Metastatic Urothelial Carcinoma After Previous Cisplatin-based Chemotherapy for Localized Disease: A Retrospective Comparison of Different Chemotherapy Regimens. Clin Genitourin Cancer 2021; 19:125-134. [DOI: 10.1016/j.clgc.2020.10.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 10/27/2020] [Accepted: 10/30/2020] [Indexed: 11/27/2022]
|
68
|
Botticelli A, Cirillo A, Pomati G, Cerbelli B, Scagnoli S, Roberto M, Gelibter A, Mammone G, Calandrella ML, Cerbelli E, Di Pietro FR, De Galitiis F, Lanzetta G, Cortesi E, Mezi S, Marchetti P. The role of opioids in cancer response to immunotherapy. J Transl Med 2021; 19:119. [PMID: 33757546 PMCID: PMC7988927 DOI: 10.1186/s12967-021-02784-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 03/11/2021] [Indexed: 02/08/2023] Open
Abstract
Background The response to immunotherapy can be impaired by several factors including external intervention such as drug interactions with immune system. We aimed to examine the immunomodulatory action of opioids, since immune cells express opioid receptors able to negatively influence their activities. Methods This observational, multicenter, retrospective study, recruited patients with different metastatic solid tumors, who have received immunotherapy between September 2014 and September 2019. Immunotherapy was administered according to the standard schedule approved for each primary tumor and line of treatment. The concomitant intake of antibiotics, antifungals, corticosteroids and opioids were evaluated in all included patients. The relationship between tumor response to immunotherapy and the oncological outcomes were evaluated. A multivariate Cox-proportional hazard model was used to identify independent prognostic factors for survival. Results One hundred ninety-three patients were recruited. Overall, progression-free survival (PFS) and overall survival (OS) were significantly shorter in those patients taking opioids than in those who didn’t (median PFS, 3 months vs. 19 months, HR 1.70, 95% CI 1.37–2.09, p < 0.0001; median OS, 4 months vs. 35 months, HR 1.60, 95% CI 1.26–2.02, p < 0.0001). In addition, PFS and OS were significantly impaired in those patients taking corticosteroids, antibiotics or antifungals, in those patients with an ECOG PS ≥ 1 and in patients with a high tumor burden. Using the multivariate analyses, opioids and ECOG PS were independent prognostic factors for PFS, whereas only ECOG PS resulted to be an independent prognostic factor for OS, with trend toward significance for opioids as well as tumor burden. Discussion Our study suggests that the concomitant administration of drugs as well as some clinical features could negatively predict the outcomes of cancer patients receiving immunotherapy. In particular, opioids use during immunotherapy is associated with early progression, potentially representing a predictive factor for PFS and negatively influencing OS as well. Conclusions A possible negative drug interaction able to impair the immune response to anti-PD-1/PD-L1 agents has been highlighted. Our findings suggest the need to further explore the impact of opioids on immune system modulation and their role in restoring the response to immunotherapy treatment, thereby improving patients' outcomes.
Collapse
Affiliation(s)
- Andrea Botticelli
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, 00185, Rome, Italy
| | - Alessio Cirillo
- Department of Radiological, Oncological and Pathological Science, Sapienza University of Rome, 00185, Rome, Italy
| | - Giulia Pomati
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161, Rome, Italy.
| | - Bruna Cerbelli
- Department of Radiological, Oncological and Pathological Science, Sapienza University of Rome, 00185, Rome, Italy
| | - Simone Scagnoli
- Department of Medical and Surgical Sciences and Translational Medicine, University of Rome Sapienza, 00185, Rome, Italy
| | - Michela Roberto
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, 00185, Rome, Italy
| | - Alain Gelibter
- Department of Radiological, Oncological and Pathological Science, Sapienza University of Rome, 00185, Rome, Italy
| | - Giulia Mammone
- Department of Radiological, Oncological and Pathological Science, Sapienza University of Rome, 00185, Rome, Italy
| | - Maria Letizia Calandrella
- Department of Radiological, Oncological and Pathological Science, Sapienza University of Rome, 00185, Rome, Italy
| | - Edoardo Cerbelli
- Department of Radiological, Oncological and Pathological Science, Sapienza University of Rome, 00185, Rome, Italy
| | | | | | - Gaetano Lanzetta
- Medical Oncology Unit, Italian Neuro-Traumatology Institute, 00046, Grottaferrata, Italy
| | - Enrico Cortesi
- Department of Radiological, Oncological and Pathological Science, Sapienza University of Rome, 00185, Rome, Italy
| | - Silvia Mezi
- Department of Radiological, Oncological and Pathological Science, Sapienza University of Rome, 00185, Rome, Italy
| | - Paolo Marchetti
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, 00185, Rome, Italy
| |
Collapse
|
69
|
Waninger JJ, Ma VT, Journey S, Skvarce J, Chopra Z, Tezel A, Bryant AK, Mayo C, Sun Y, Sankar K, Ramnath N, Lao C, Sussman JB, Fecher L, Alva A, Green MD. Validation of the American Joint Committee on Cancer Eighth Edition Staging of Patients With Metastatic Cutaneous Melanoma Treated With Immune Checkpoint Inhibitors. JAMA Netw Open 2021; 4:e210980. [PMID: 33687443 PMCID: PMC7944385 DOI: 10.1001/jamanetworkopen.2021.0980] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
IMPORTANCE Immune checkpoint inhibitors (ICIs) have transformed the survival of patients with metastatic melanoma. Patient prognosis is reflected by the American Joint Committee on Cancer (AJCC) staging system; however, it is unknown whether the metastatic (M) stage categories for cutaneous melanoma remain informative of prognosis in patients who have received ICIs. OBJECTIVES To evaluate the outcomes of patients with metastatic cutaneous melanoma based on the M stage category from the AJCC eighth edition and to determine whether these designations continue to inform the prognosis of patients who have received ICIs. DESIGN, SETTING, AND PARTICIPANTS This cohort study included patients with metastatic cutaneous melanoma who were treated between August 2006 and August 2019 at the University of Michigan. The estimated median follow-up time was 35.5 months. Patient data were collected via the electronic medical record system. Critical findings were externally validated in a multicenter nationwide cohort of patients treated within the Veterans Affairs health care system. Data analysis was conducted from February 2020 to January 2021. EXPOSURES All patients were treated with dual-agent concurrent ipilimumab and nivolumab followed by maintenance nivolumab or single-agent ipilimumab, nivolumab, or pembrolizumab therapy. Patients were staged using the AJCC eighth edition. MAIN OUTCOMES AND MEASURES Univariable and multivariable analyses were used to assess the prognostic value of predefined clinicopathologic baseline factors on survival. RESULTS In a discovery cohort of 357 patients (mean [SD] age, 62.6 [14.2] years; 254 [71.1%] men) with metastatic cutaneous melanoma treated with ICIs, the M category in the AJCC eighth edition showed limited prognostic stratification by both univariable and multivariable analyses. The presence of liver metastases and elevated levels of serum lactate dehydrogenase (LDH) offered superior prognostic separation compared with the M category (liver metastases: hazard ratio, 2.22; 95% CI, 1.48-3.33; P < .001; elevated serum LDH: hazard ratio, 1.73; 95% CI, 1.16-2.58; P = .007). An updated staging system based on these factors was externally validated in a cohort of 652 patients (mean [SD] age, 67.9 [11.6] years; 630 [96.6%] men), with patients without liver metastases or elevated LDH levels having the longest survival (median overall survival, 30.7 months). CONCLUSIONS AND RELEVANCE This study found that the AJCC eighth edition M category was poorly reflective of prognosis in patients receiving ICIs. Future staging systems could consider emphasizing the presence of liver metastases and elevated LDH levels. Additional studies are needed to confirm the importance of these and other prognostic biomarkers.
Collapse
Affiliation(s)
- Jessica J. Waninger
- University of Michigan Medical School, University of Michigan, Ann Arbor
- Department of Cellular and Molecular Biology, University of Michigan, Ann Arbor
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor
| | - Vincent T. Ma
- Division of Hematology Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor
| | - Sara Journey
- University of Michigan Medical School, University of Michigan, Ann Arbor
| | - Jeremy Skvarce
- University of Michigan Medical School, University of Michigan, Ann Arbor
| | - Zoey Chopra
- University of Michigan Medical School, University of Michigan, Ann Arbor
| | - Alangoya Tezel
- University of Michigan Medical School, University of Michigan, Ann Arbor
| | - Alex K. Bryant
- Department of Radiation Oncology, University of Michigan, Ann Arbor
| | - Charles Mayo
- Department of Radiation Oncology, University of Michigan, Ann Arbor
| | - Yilun Sun
- Department of Radiation Oncology, University of Michigan, Ann Arbor
- Department of Biostatistics, University of Michigan, Ann Arbor
| | - Kamya Sankar
- Rogel Cancer Center, University of Michigan, Ann Arbor
| | - Nithya Ramnath
- Rogel Cancer Center, University of Michigan, Ann Arbor
- Department of Hematology Oncology, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan
| | - Christopher Lao
- Division of Hematology Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor
- Rogel Cancer Center, University of Michigan, Ann Arbor
| | - Jeremy B. Sussman
- Department of Medicine, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan
- Center for Clinical Management Research, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan
- Institute for Healthcare Policy and Innovation, University of Michigan, Ann Arbor
| | - Leslie Fecher
- Division of Hematology Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor
- Rogel Cancer Center, University of Michigan, Ann Arbor
| | - Ajjai Alva
- Division of Hematology Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor
- Department of Hematology Oncology, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan
| | - Michael D. Green
- Department of Radiation Oncology, University of Michigan, Ann Arbor
- Rogel Cancer Center, University of Michigan, Ann Arbor
- Department of Radiation Oncology, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan
| |
Collapse
|
70
|
Parikh AB, Zhong X, Mellgard G, Qin Q, Patel VG, Wang B, Alerasool P, Garcia P, Leiter A, Gallagher EJ, Clinton S, Mortazavi A, Monk P, Folefac E, Yin M, Yang Y, Galsky M, Oh WK, Tsao CK. Risk Factors for Emergency Room and Hospital Care Among Patients With Solid Tumors on Immune Checkpoint Inhibitor Therapy. Am J Clin Oncol 2021; 44:114-120. [PMID: 33417323 PMCID: PMC7902456 DOI: 10.1097/coc.0000000000000793] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
OBJECTIVES Immune checkpoint inhibitors (ICIs) are being increasingly used across cancer types. Emergency room (ER) and inpatient (IP) care, common in patients with cancer, remain poorly defined in this specific population, and risk factors for such care are unknown. METHODS We retrospectively reviewed charts for patients with solid tumors who received >1 ICI dose at 1 of 2 sites from January 1, 2011 to April 28, 2017. Demographics, medical history, cancer diagnosis/therapy/toxicity details, and outcomes were recorded. Descriptive data detailing ER/IP care at the 2 associated hospitals during ICI therapy (from first dose to 3 mo after last dose) were collected. The Fisher exact test and multivariate regression analysis was used to study differences between patients with versus without ER/IP care during ICI treatment. RESULTS Among 345 patients studied, 50% had at least 1 ER visit during ICI treatment and 43% had at least 1 IP admission. Six percent of ER/IP visits eventually required intensive care. A total of 12% of ER/IP visits were associated with suspected or confirmed immune-related adverse events. Predictors of ER care were African-American race (odds ratio [OR]: 3.83, P=0.001), Hispanic ethnicity (OR: 3.12, P=0.007), and coronary artery disease (OR: 2.43, P=0.006). Predictors of IP care were African-American race (OR: 2.38, P=0.024), Hispanic ethnicity (OR: 2.29, P=0.045), chronic kidney disease (OR: 3.89, P=0.006), angiotensin converting enzyme inhibitor/angiotensin receptor blocker medication use (OR: 0.44, P=0.009), and liver metastasis (OR: 2.32, P=0.003). CONCLUSIONS Understanding demographic and clinical risk factors for ER/IP care among patients on ICIs can help highlight disparities, prospectively identify high-risk patients, and inform preventive programs aimed at reducing such care.
Collapse
Affiliation(s)
- Anish B Parikh
- Genitourinary Oncology Section, Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center – James Cancer Hospital, Columbus OH USA
- Tisch Cancer Institute, Division of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York NY USA
| | - Xiaobo Zhong
- Division of Biostatistics, Icahn School of Medicine at Mount Sinai, New York NY USA
| | | | - Qian Qin
- Tisch Cancer Institute, Division of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York NY USA
| | - Vaibhav G Patel
- Tisch Cancer Institute, Division of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York NY USA
| | - Bo Wang
- Tisch Cancer Institute, Division of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York NY USA
| | - Parissa Alerasool
- Tisch Cancer Institute, Division of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York NY USA
- New York Medical College, Valhalla NY USA
| | - Philip Garcia
- Icahn School of Medicine at Mount Sinai, New York NY USA
| | - Amanda Leiter
- Division of Endocrinology, Diabetes, and Bone Disease, Icahn School of Medicine at Mount Sinai, New York NY USA
| | - Emily J Gallagher
- Tisch Cancer Institute, Division of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York NY USA
- Division of Endocrinology, Diabetes, and Bone Disease, Icahn School of Medicine at Mount Sinai, New York NY USA
| | - Steven Clinton
- Genitourinary Oncology Section, Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center – James Cancer Hospital, Columbus OH USA
| | - Amir Mortazavi
- Genitourinary Oncology Section, Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center – James Cancer Hospital, Columbus OH USA
| | - Paul Monk
- Genitourinary Oncology Section, Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center – James Cancer Hospital, Columbus OH USA
| | - Edmund Folefac
- Genitourinary Oncology Section, Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center – James Cancer Hospital, Columbus OH USA
| | - Ming Yin
- Genitourinary Oncology Section, Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center – James Cancer Hospital, Columbus OH USA
| | - Yuanquan Yang
- Genitourinary Oncology Section, Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center – James Cancer Hospital, Columbus OH USA
| | - Matthew Galsky
- Tisch Cancer Institute, Division of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York NY USA
| | - William K Oh
- Tisch Cancer Institute, Division of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York NY USA
| | - Che-Kai Tsao
- Tisch Cancer Institute, Division of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York NY USA
| |
Collapse
|
71
|
Ma VT, Su CT, Hu M, Taylor JMG, Daignault-Newton S, Kellezi O, Dahl MN, Shah MA, Erickson S, Lora J, Hamasha R, Ali A, Yancey S, Kiros L, Balicki HM, Winfield DC, Green MD, Alva AS. Characterization of outcomes in patients with advanced genitourinary malignancies treated with immune checkpoint inhibitors. Urol Oncol 2021; 39:437.e1-437.e9. [PMID: 33495117 DOI: 10.1016/j.urolonc.2021.01.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 12/13/2020] [Accepted: 01/04/2021] [Indexed: 12/11/2022]
Abstract
PURPOSE Several immune checkpoint inhibitors (ICIs) are FDA approved for treatment of genitourinary (GU) malignancies. We aim to determine demographic and clinicopathologic characteristics that significantly affect clinical outcomes in patients with advanced stage GU malignancies treated with ICIs. MATERIALS AND METHODS We performed a single-center, consecutive, retrospective cohort analysis on patients with metastatic or unresectable GU malignancies who were treated with ICIs at the University of Michigan. Immune-related adverse events (irAEs), putative immune-mediated allergies, and overall response rates (ORR) were assessed. Comorbidity index scores were calculated. Survival analysis was performed to evaluate progression-free survival (PFS) and overall survival (OS), stratifying and controlling for a variety of clinicopathologic baseline factors including site of metastases. RESULTS A total of 160 patients were identified with advanced renal cell carcinoma (RCC) or urothelial carcinoma. Median PFS and OS were 5.0 and 23.6 months for RCC, and 2.8 and 9.6 months for urothelial carcinoma, respectively. Patients who experienced increased frequency and higher grade irAEs had better ICI treatment response (P < 0.0001). Presence of liver metastases was associated with poor response to ICI therapy (P = 0.001). Multivariable modeling demonstrates that patients with urothelial carcinoma and liver metastases had statistically worse PFS and OS compared to patients with RCC or other sites of metastases, respectively. CONCLUSION Greater frequency and higher grades of irAEs are associated with better treatment response in patients with RCC and urothelial malignancy receiving ICI therapy. The presence of liver metastases denotes a negative predictive marker for immunotherapy efficacy. SUMMARY Immune checkpoint inhibitors (ICI) are increasingly used to treat genitourinary (GU) malignancies. However, clinical data regarding patients with advanced-stage GU malignancies treated with ICI is lacking. Thus, we performed a single-center, retrospective cohort study on patients with metastatic and unresectable renal cell carcinoma (RCC) and urothelial carcinoma who were treated with ICIs at the University of Michigan to provide demographic and clinicopathologic data regarding this population. We specifically focused on immune-related adverse events (irAEs), immune-mediated allergies, and the associated overall response rates (ORR). To better assess performance status, we calculated comorbidity scores for all patients. Finally, survival analyses for progression-free survival (PFS) and overall survival (OS) were performed using Kaplan-Meier analysis and Cox proportional hazards modeling, stratifying and controlling for clinicopathologic baseline factors, including sites of metastases, in our multivariable analysis. A total of 160 patients were identified with advanced RCC or urothelial carcinoma. We found decreased PFS (2.8 vs. 5.0 months) and decreased OS (9.8 vs. 23.6 months) for urothelial carcinoma compared to RCC patients. We noted that patients who experienced increased frequency and higher grades of irAEs had better treatment ORR with ICI therapy (P ≤ 0.0001). The presence of liver metastases was associated with worse ORR (P = 0.001), PFS (P = 0.0014), and OS (P = 0.0028) compared to other sites of metastases including lymph node, lung, and CNS/bone. The poor PFS and OS associated with urothelial carcinoma and liver metastases were preserved in our multivariable modeling after controlling for pertinent clinical factors. We conclude that greater frequency and higher grades of irAEs are associated with better treatment response in GU malignancy patients receiving ICI, a finding that is consistent with published studies in other cancers. The presence of liver metastases represents a significantly poor predictive marker in GU malignancy treated with ICI. Our findings contribute to the growing body of literature that seeks to understand the clinicopathologic variables and outcomes associated with ICI therapy.
Collapse
Affiliation(s)
- Vincent T Ma
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, MI.
| | - Christopher T Su
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, MI
| | - Miriam Hu
- Department of Biostatistics, University of Michigan, Ann Arbor, MI
| | | | | | - Olesia Kellezi
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, MI
| | - Megan N Dahl
- College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI
| | - Miloni A Shah
- College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI
| | - Stephanie Erickson
- Department of PreMedical PostBaccalaureate, University of Michigan, Ann Arbor, MI
| | - Jessica Lora
- College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI
| | - Reema Hamasha
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, MI
| | - Alicia Ali
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, MI
| | - Sabrina Yancey
- College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI
| | - Leah Kiros
- College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI
| | - Hannah M Balicki
- College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, MI
| | - Daniel C Winfield
- Department of PreMedical PostBaccalaureate, University of Michigan, Ann Arbor, MI
| | - Michael D Green
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI
| | - Ajjai S Alva
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, MI
| |
Collapse
|
72
|
Skribek M, Rounis K, Afshar S, Grundberg O, Friesland S, Tsakonas G, Ekman S, De Petris L. Effect of corticosteroids on the outcome of patients with advanced non-small cell lung cancer treated with immune-checkpoint inhibitors. Eur J Cancer 2021; 145:245-254. [PMID: 33419647 DOI: 10.1016/j.ejca.2020.12.012] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 11/23/2020] [Accepted: 12/14/2020] [Indexed: 01/21/2023]
Abstract
INTRODUCTION We analysed patients with advanced non-small cell lung cancer (NSCLC) who were treated with immune-checkpoint inhibitors (ICIs) to address the effect of the timeline and reason for corticosteroid administration on survival outcomes. METHODS We retrospectively collected clinical data of non-oncogenic driven, advanced NSCLC patients treated with ICIs at Karolinska University Hospital, including the timeline and reason for steroid administration. Steroid administration was defined as > 10 mg prednisolone equivalent for ≥10 days. We subcategorized patients based on the aetiology of steroid administration into three subgroups: a) steroids for supportive reasons but not for cancer palliation; b) steroids for the palliation of cancer-related symptoms; c) steroids for the management of immune-related adverse events (irAEs). Furthermore, to analyse the timeline, patients were categorised into two groups; those who received corticosteroids within 2 weeks before until 2 days after ICI initiation and those who received steroids later during their treatment course. RESULTS Analysed data from 196 patients showed 46.3% of patients received corticosteroids. Steroid administration due to irAEs did not affect overall survival (OS) (p = 0.38) compared with the steroid naïve group. Only steroid administration for the palliation of cancer-related symptoms was an independent predictor for shorter OS (HR = 2.7; 95% CI, 1.5-4.9). The timeline of steroid administration did not affect OS (p = 0.456) in our cohort. CONCLUSIONS Steroids due to irAEs do not appear to hamper ICI efficacy. However, the administration of high-dose steroids to palliate malignancy-associated symptoms might reflect the dismal prognosis of this patient group.
Collapse
Affiliation(s)
- Marcus Skribek
- Thoracic Oncology Center, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden; Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Konstantinos Rounis
- Thoracic Oncology Center, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden; Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden; Department of Medical Oncology, University General Hospital, Heraklion, Crete, Greece
| | - Soren Afshar
- Thoracic Oncology Center, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
| | - Oscar Grundberg
- Thoracic Oncology Center, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden; Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Signe Friesland
- Thoracic Oncology Center, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden; Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Georgios Tsakonas
- Thoracic Oncology Center, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden; Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Simon Ekman
- Thoracic Oncology Center, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden; Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Luigi De Petris
- Thoracic Oncology Center, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden; Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
73
|
Lombard A, Mistry H, Aarons L, Ogungbenro K. Dose individualisation in oncology using chemotherapy-induced neutropenia: Example of docetaxel in non-small cell lung cancer patients. Br J Clin Pharmacol 2020; 87:2053-2063. [PMID: 33075149 DOI: 10.1111/bcp.14614] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 09/25/2020] [Accepted: 10/09/2020] [Indexed: 11/28/2022] Open
Abstract
AIMS Chemotherapy-induced neutropenia has been associated with an increase in overall survival in non-small cell lung cancer patients. Therefore, neutrophil counts could be an interesting biomarker for drug efficacy as well as linked directly to toxicity. For drugs where neutropenia is dose limiting, neutrophil counts might be used for monitoring drug effect and for dosing optimisation. METHODS The relationship between drug effect on the first cycle neutrophil counts and patient survival was explored in a Phase III clinical trial where patients with non-small cell lung cancer were treated with docetaxel. Once the association has been established, dosing optimisation was performed for patients with severe toxicities (neutropenia) without compromising drug efficacy (overall survival). RESULTS After taking into account baseline prognostic factors, such as Eastern Cooperative Oncology Group performance status, smoking status, liver metastasis, tumour burden, neutrophil counts and albumin levels, a model-predicted drug effect on the first cycle neutrophil counts was strongly associated with patient survival (P = .005). Utilising this relationship in a dose optimisation algorithm, 194 out of 366 patients would have benefited from a dose reduction after the first cycle of docetaxel. The simulated 1-year survival probabilities associated with the original dose and the individualised dose were not different. CONCLUSION The strong relationship between drug effect on the first cycle neutrophil counts and patient survival suggests that this variable could be used to individualise dosing, possibly without needing pharmacokinetic samples. The algorithm highlights that doses could be reduced in case of severe haematological toxicities without compromising drug efficacy.
Collapse
Affiliation(s)
- Aurélie Lombard
- Centre for Applied Pharmacokinetic Research, University of Manchester, UK.,Division of Pharmacy and Optometry, University of Manchester, UK
| | - Hitesh Mistry
- Division of Pharmacy and Optometry, University of Manchester, UK.,Division of Cancer Sciences, University of Manchester, UK
| | - Leon Aarons
- Centre for Applied Pharmacokinetic Research, University of Manchester, UK.,Division of Pharmacy and Optometry, University of Manchester, UK
| | - Kayode Ogungbenro
- Centre for Applied Pharmacokinetic Research, University of Manchester, UK.,Division of Pharmacy and Optometry, University of Manchester, UK
| |
Collapse
|
74
|
Bohlok A, Vermeulen P, Leduc S, Latacz E, Botzenhart L, Richard F, De Schepper M, Geukens T, Lucidi V, Ignatiadis M, Aftimos P, Sotiriou C, Piccart M, Hendlisz A, Van Laere S, Dirix L, Noël JC, Biganzoli E, Larsimont D, Desmedt C, Donckier V. Association between the histopathological growth patterns of liver metastases and survival after hepatic surgery in breast cancer patients. NPJ Breast Cancer 2020; 6:64. [PMID: 33339824 PMCID: PMC7749172 DOI: 10.1038/s41523-020-00209-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 11/12/2020] [Indexed: 12/16/2022] Open
Abstract
Currently, there are no markers to identify patients with liver-only or liver-dominant metastases that would benefit from hepatic surgery. Here we characterized histopathological growth patterns (HGPs) of liver metastases in a consecutive series of 36 breast cancer patients who underwent hepatic surgery. Survival analyses showed that the presence of a desmoplastic HGP in the liver metastases (a rim of fibrous tissue separating cancer cells from the liver parenchyma, present in 20 (56%) patients) is independently associated with favorable progression-free and overall survival when compared with the replacement HGP (cancer cells growing into the liver parenchyma, present in 16 (44%) patients).
Collapse
Affiliation(s)
- Ali Bohlok
- Department of Surgery, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Peter Vermeulen
- Translational Cancer Research Unit, GZA Hospitals & CORE, MIPRO, University of Antwerp, Antwerp, Belgium
- Department of Oncological Research, Oncology Center, GZA Hospitals Sint-Augustinus, Antwerp, Belgium
| | - Sophia Leduc
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Emily Latacz
- Translational Cancer Research Unit, GZA Hospitals & CORE, MIPRO, University of Antwerp, Antwerp, Belgium
| | - Lara Botzenhart
- Department of Oncological Research, Oncology Center, GZA Hospitals Sint-Augustinus, Antwerp, Belgium
| | - François Richard
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Maxim De Schepper
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Tatjana Geukens
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Valerio Lucidi
- Department of Abdominal Surgery, Hôpital Erasme, Université Libre de Bruxelles, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Michail Ignatiadis
- Medical Oncology Clinic, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Philippe Aftimos
- Clinical Trials Conduct Unit, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Christos Sotiriou
- Breast Cancer Translational Lab, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Martine Piccart
- Medical Oncology Clinic, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Alain Hendlisz
- Medical Oncology Department, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Steven Van Laere
- Translational Cancer Research Unit, GZA Hospitals & CORE, MIPRO, University of Antwerp, Antwerp, Belgium
| | - Luc Dirix
- Translational Cancer Research Unit, GZA Hospitals & CORE, MIPRO, University of Antwerp, Antwerp, Belgium
- Department of Oncological Research, Oncology Center, GZA Hospitals Sint-Augustinus, Antwerp, Belgium
| | - Jean-Christophe Noël
- Department of Pathology, Hôpital Erasme, Université Libre de Bruxelles, Brussels, Belgium
| | - Elia Biganzoli
- Department of Clinical Sciences and Community Health & DSRC, University of Milan, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Denis Larsimont
- Department of Pathology, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Christine Desmedt
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Leuven, Belgium.
| | - Vincent Donckier
- Department of Surgery, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium.
| |
Collapse
|
75
|
Wei XL, Luo X, Sheng H, Wang Y, Chen DL, Li JN, Wang FH, Xu RH. PD-L1 expression in liver metastasis: its clinical significance and discordance with primary tumor in colorectal cancer. J Transl Med 2020; 18:475. [PMID: 33308232 PMCID: PMC7730753 DOI: 10.1186/s12967-020-02636-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Accepted: 11/27/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The outcomes of immune checkpoint inhibitors in cancer patients with liver metastases are poor, which may be related to a different tumor microenvironment in liver metastases from primary tumors. This study was aimed to analyze PD-L1 expression and the immune microenvironment status in liver metastases and compare the differences of PD-L1 expression between primary tumors and liver metastases of colorectal cancer. METHODS 74 cases of pathologically confirmed colorectal cancer with liver metastasis underwent resection from our hospital were included. Tissue microarrays were used for the interpretation of PD-L1 expression, cluster of differentiation 4 (CD4) and CD8 density by immunohistochemistry. We evaluated the disparity between primary tumor and liver metastasis in PD-L1 expression, CD4 and CD8 density and analyzed the factors associated with obvious PD-L1 disparity. RESULTS The expression of PD-L1 was positively related to the density of CD4 and CD8 in liver metastases. The expression of PD-L1 in liver metastases was higher than in primary tumors in certain subgroups, including patients with concurrent liver metastases (n = 63, p = 0.05), patients receiving concurrent resection of primary and metastatic tumors (n = 56, p = 0.04). The two subgroups generally reflected those without inconsistent external influences, such as treatment and temporal factors, between primary tumors and liver metastases. In these subgroups, the intrinsic differences of microenvironment between primary tumors and liver metastases could be identified. Furthermore, tumor differentiation [moderate vs. poor: OR = 0.23, 95% CI: 0.03-0.99, p = 0.05)] were demonstrated to be associated with obvious discordance of PD-L1 expression between primary tumors and liver metastases. CONCLUSIONS The expression of PD-L1 in liver metastases was higher than in primary tumors in subgroups, reflecting intrinsic microenvironment differences between primary and metastatic tumors. Obvious discordance of PD-L1 expression between primary tumor and liver metastasis was significantly related to the tumor differentiation.
Collapse
Affiliation(s)
- Xiao-Li Wei
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dong Feng Road East, Guangzhou, 510060, Guangdong, China
| | - Xuan Luo
- Department of Hepatobiliary Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Hui Sheng
- State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Yun Wang
- Department of Hematologic Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Dong-Liang Chen
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dong Feng Road East, Guangzhou, 510060, Guangdong, China
| | - Jia-Ning Li
- Department of Clinical Trial Center, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Feng-Hua Wang
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dong Feng Road East, Guangzhou, 510060, Guangdong, China.
| | - Rui-Hua Xu
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dong Feng Road East, Guangzhou, 510060, Guangdong, China.
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou, 510060, China.
| |
Collapse
|
76
|
An Update on the Role of Talimogene Laherparepvec (T-VEC) in the Treatment of Melanoma: Best Practices and Future Directions. Am J Clin Dermatol 2020; 21:821-832. [PMID: 32767272 DOI: 10.1007/s40257-020-00554-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Talimogene laherparepvec (T-VEC) is the first agent approved for cancer in the emerging class of oncolytic viral therapies. While T-VEC was approved for the treatment of advanced melanoma in 2015, clinical utilization has been hampered by rapid changes in the therapeutic landscape of melanoma related to advances in both immune checkpoint blockade and targeted therapy, cumbersome logistics involved in T-VEC administration, biosafety concerns, and a perception that T-VEC has limited impact on uninjected, visceral disease. However, with further survival follow-up from the phase III OPTiM (OncovexGM-CSF Pivotal Trial in Melanoma), along with new real-world data and consensus guidelines on safe administration of oncolytic viruses, a roadmap for when and how to use T-VEC has been emerging. In addition, preliminary data have demonstrated improved therapeutic responses to T-VEC in combination with immune checkpoint blockade in patients with melanoma without additive toxicity. This review provides an update on recent data with T-VEC alone and in combination with other agents. The emerging data provide guidance for how to better utilize T-VEC for patients with melanoma and identifies critical areas for clinical investigation to expand the role of T-VEC in combination strategies for the treatment of melanoma and perhaps other cancers.
Collapse
|
77
|
Hack SP, Zhu AX, Wang Y. Augmenting Anticancer Immunity Through Combined Targeting of Angiogenic and PD-1/PD-L1 Pathways: Challenges and Opportunities. Front Immunol 2020; 11:598877. [PMID: 33250900 PMCID: PMC7674951 DOI: 10.3389/fimmu.2020.598877] [Citation(s) in RCA: 160] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 10/08/2020] [Indexed: 12/11/2022] Open
Abstract
Cancer immunotherapy (CIT) with antibodies targeting the programmed cell death 1 protein (PD-1)/programmed cell death 1 ligand 1 (PD-L1) axis have changed the standard of care in multiple cancers. However, durable antitumor responses have been observed in only a minority of patients, indicating the presence of other inhibitory mechanisms that act to restrain anticancer immunity. Therefore, new therapeutic strategies targeted against other immune suppressive mechanisms are needed to enhance anticancer immunity and maximize the clinical benefit of CIT in patients who are resistant to immune checkpoint inhibition. Preclinical and clinical studies have identified abnormalities in the tumor microenvironment (TME) that can negatively impact the efficacy of PD-1/PD-L1 blockade. Angiogenic factors such as vascular endothelial growth factor (VEGF) drive immunosuppression in the TME by inducing vascular abnormalities, suppressing antigen presentation and immune effector cells, or augmenting the immune suppressive activity of regulatory T cells, myeloid-derived suppressor cells, and tumor-associated macrophages. In turn, immunosuppressive cells can drive angiogenesis, thereby creating a vicious cycle of suppressed antitumor immunity. VEGF-mediated immune suppression in the TME and its negative impact on the efficacy of CIT provide a therapeutic rationale to combine PD-1/PD-L1 antibodies with anti-VEGF drugs in order to normalize the TME. A multitude of clinical trials have been initiated to evaluate combinations of a PD-1/PD-L1 antibody with an anti-VEGF in a variety of cancers. Recently, the positive results from five Phase III studies in non-small cell lung cancer (adenocarcinoma), renal cell carcinoma, and hepatocellular carcinoma have shown that combinations of PD-1/PD-L1 antibodies and anti-VEGF agents significantly improved clinical outcomes compared with respective standards of care. Such combinations have been approved by health authorities and are now standard treatment options for renal cell carcinoma, non-small cell lung cancer, and hepatocellular carcinoma. A plethora of other randomized studies of similar combinations are currently ongoing. Here, we discuss the principle mechanisms of VEGF-mediated immunosuppression studied in preclinical models or as part of translational clinical studies. We also discuss data from recently reported randomized clinical trials. Finally, we discuss how these concepts and approaches can be further incorporated into clinical practice to improve immunotherapy outcomes for patients with cancer.
Collapse
Affiliation(s)
- Stephen P. Hack
- Product Development (Oncology), Genentech, Inc., South San Francisco, CA, United States
| | - Andrew X. Zhu
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, United States
- Jiahui International Cancer Center, Jiahui Health, Shanghai, China
| | - Yulei Wang
- Product Development (Oncology), Genentech, Inc., South San Francisco, CA, United States
| |
Collapse
|
78
|
Lee JC, Mehdizadeh S, Smith J, Young A, Mufazalov IA, Mowery CT, Daud A, Bluestone JA. Regulatory T cell control of systemic immunity and immunotherapy response in liver metastasis. Sci Immunol 2020; 5:eaba0759. [PMID: 33008914 PMCID: PMC7755924 DOI: 10.1126/sciimmunol.aba0759] [Citation(s) in RCA: 179] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 09/09/2020] [Indexed: 12/18/2022]
Abstract
Patients with cancer with liver metastasis demonstrate significantly worse outcomes than those without liver metastasis when treated with anti-PD-1 immunotherapy. The mechanism of liver metastases-induced reduction in systemic antitumor immunity is unclear. Using a dual-tumor immunocompetent mouse model, we found that the immune response to tumor antigen presence within the liver led to the systemic suppression of antitumor immunity. The immune suppression was antigen specific and associated with the coordinated activation of regulatory T cells (Tregs) and modulation of intratumoral CD11b+ monocytes. The dysfunctional immune state could not be reversed by anti-PD-1 monotherapy unless Treg cells were depleted (anti-CTLA-4) or destabilized (EZH2 inhibitor). Thus, this study provides a mechanistic understanding and rationale for adding Treg and CD11b+ monocyte targeting agents in combination with anti-PD-1 to treat patients with cancer with liver metastasis.
Collapse
MESH Headings
- Animals
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- CD11b Antigen/metabolism
- CD8-Positive T-Lymphocytes/drug effects
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- CTLA-4 Antigen/antagonists & inhibitors
- CTLA-4 Antigen/metabolism
- Cell Line, Tumor/transplantation
- Disease Models, Animal
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/immunology
- Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors
- Enhancer of Zeste Homolog 2 Protein/metabolism
- Female
- Humans
- Immune Checkpoint Inhibitors/pharmacology
- Immune Checkpoint Inhibitors/therapeutic use
- Liver Neoplasms/drug therapy
- Liver Neoplasms/immunology
- Liver Neoplasms/secondary
- Lymphocyte Depletion/methods
- Lymphocytes, Tumor-Infiltrating/drug effects
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Male
- Mice
- Mice, Transgenic
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Tumor Escape/drug effects
- Tumor Microenvironment/drug effects
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- James C Lee
- Division of Hematology and Oncology, University of California, San Francisco, San Francisco, CA 94143, USA.
- Sean N. Parker Autoimmune Research Laboratory, University of California, San Francisco, San Francisco, CA 94143, USA
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Sadaf Mehdizadeh
- Sean N. Parker Autoimmune Research Laboratory, University of California, San Francisco, San Francisco, CA 94143, USA
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jennifer Smith
- Sean N. Parker Autoimmune Research Laboratory, University of California, San Francisco, San Francisco, CA 94143, USA
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Arabella Young
- Sean N. Parker Autoimmune Research Laboratory, University of California, San Francisco, San Francisco, CA 94143, USA
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
- QIMR Berghofer Medical Research Institute, Herston, Queensland 4006, Australia
| | - Ilgiz A Mufazalov
- Sean N. Parker Autoimmune Research Laboratory, University of California, San Francisco, San Francisco, CA 94143, USA
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Cody T Mowery
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
- Medical Scientist Training Program, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Adil Daud
- Division of Hematology and Oncology, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94158, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA 94129, USA
| | - Jeffrey A Bluestone
- Sean N. Parker Autoimmune Research Laboratory, University of California, San Francisco, San Francisco, CA 94143, USA.
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94158, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA 94129, USA
| |
Collapse
|
79
|
Choi YJ, Kim T, Kim EY, Lee SH, Kwon DS, Chang YS. Prediction model for hyperprogressive disease in non-small cell lung cancer treated with immune checkpoint inhibitors. Thorac Cancer 2020; 11:2793-2803. [PMID: 32779394 PMCID: PMC7529559 DOI: 10.1111/1759-7714.13594] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 07/08/2020] [Accepted: 07/09/2020] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Hyperprogressive disease (HPD) is a paradoxical acceleration of tumor growth after immune checkpoint inhibitor (ICI) treatment. This study aimed to identify the risk factors and to present a predictive model for HPD in patients treated with ICIs. METHODS A total of 78 non-small cell lung cancer (NSCLC) cases, treated with at least two cycles of ICIs who underwent computed tomography (CT) for response assessment were recruited into the study from January 2016 to August 2019. HPD was defined by the following criteria: (i) time-to-treatment failure <2 months; (ii) a 50% increase in the sum of target lesion diameters; (iii) new development of at least two lesions in an already involved organ; (iv) appearance of a new organ lesion; and (v) a decrease in ECOG PS 2. RESULTS Of the 78 total patients, 15 (19.2%) had HPD. The risk factors of HPD were age; primary lesion size; and metastases in the contralateral lung, pleura, liver, and bone in multivariable logistic regression (odds ratio [OR]; 0.9038, 1.6619, 28.5913, 23.8264, 14.5711, and 20.1533, respectively, all P-values < 0.05). By using these risk factors, we developed a prediction model for HPD and the area under the receiver operating characteristic curve of the model was 0.9556 (95% confidence interval [CI]: 0.9133-0.9978). CONCLUSIONS HPD is relatively common and associated with a grave clinical outcome, requiring a careful monitoring in lung cancer patients treated with ICIs. Moreover, risk factors such as age, size of tumor and number of various metastatic lesions should be taken into consideration before ICI administration. KEY POINTS SIGNIFICANT FINDINGS OF THE STUDY: Age, primary lesion size, and number of metastases are risk factors of HPD. HPD is strongly associated with poor prognosis. HPD during ICI use needs comprehensive monitoring. WHAT THIS STUDY ADDS This is the first study to develop a prediction model. The area under the curve of the prediction model for HPD was 0.9556.
Collapse
Affiliation(s)
- Yong Jun Choi
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Taehee Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Eun Young Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Sang Hoon Lee
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Do Sun Kwon
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Yoon Soo Chang
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
80
|
Li X, Wang L, Chen S, Zhou F, Zhao J, Zhao W, Su C. Adverse impact of bone metastases on clinical outcomes of patients with advanced non-small cell lung cancer treated with immune checkpoint inhibitors. Thorac Cancer 2020; 11:2812-2819. [PMID: 32779372 PMCID: PMC7529562 DOI: 10.1111/1759-7714.13597] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 07/09/2020] [Accepted: 07/12/2020] [Indexed: 01/09/2023] Open
Abstract
Background Bone metastasis (BoM) is common in patients with advanced non‐small cell lung cancer (NSCLC) and considered as one of the negative prognostic factors. However, the impact of BoM on clinical outcomes of patients with advanced NSCLC treated with immune checkpoint inhibitors (ICIs) remains unclear. Methods A total of 103 patients treated with ICI monotherapy and 101 patients treated with ICIs combined with chemotherapy or antiangiogenesis therapy were retrospectively analyzed. The differences in progression‐free survival (PFS), overall survival (OS) and objective response rate (ORR) between BoM+ and BoM− were investigated. Results Of those 101 patients who received combination therapy, no significant difference between BoM− and BoM+ in terms of both median PFS and median OS (median PFS, 10.1 vs. 12.1 months, P = 0.6; median OS, NR vs. 24.6 months, P = 0.713) was determined. In contrast, of the 103 patients who received ICI monotherapy, BoM+ patients had an inferior PFS (4.2 vs. 6.7 months, P = 0.0484) and OS (12.5 vs. 23.9 months, P = 0.0036) compared with BoM− patients. The univariate and multivariate analysis in the ICI monotherapy group also identified BoM as an independent factor attenuating the efficacy of ICI monotherapy. Of all BoM+ patients who received ICI monotherapy, neither palliative radiotherapy nor bisphosphonate drugs improved OS (palliative radiotherapy: 12.5 vs. 16.7 months, P = 0.487; bisphosphonate drugs: 12.5 vs. 9.7 months, P = 0.568). Conclusions BoM attenuated the efficacy of ICI monotherapy in patients with advanced NSCLC. Of BoM+ patients who received ICI monotherapy, neither palliative radiotherapy nor bisphosphonate drugs improved OS. Other therapeutic strategies are needed for patients with BoM.
Collapse
Affiliation(s)
- Xing Li
- Medical Oncology Department, Tongji University School of Medicine Affiliated Shanghai Pulmonary Hospital, Shanghai, China.,Department of Immuno-oncology, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Lei Wang
- Medical Oncology Department, Tongji University School of Medicine Affiliated Shanghai Pulmonary Hospital, Shanghai, China
| | - Shanhao Chen
- Medical Oncology Department, Tongji University School of Medicine Affiliated Shanghai Pulmonary Hospital, Shanghai, China
| | - Fei Zhou
- Medical Oncology Department, Tongji University School of Medicine Affiliated Shanghai Pulmonary Hospital, Shanghai, China
| | - Jing Zhao
- Medical Oncology Department, Tongji University School of Medicine Affiliated Shanghai Pulmonary Hospital, Shanghai, China
| | - Wencheng Zhao
- Medical Oncology Department, Tongji University School of Medicine Affiliated Shanghai Pulmonary Hospital, Shanghai, China
| | - Chunxia Su
- Medical Oncology Department, Tongji University School of Medicine Affiliated Shanghai Pulmonary Hospital, Shanghai, China
| |
Collapse
|
81
|
Ngiow SF, Young A. Re-education of the Tumor Microenvironment With Targeted Therapies and Immunotherapies. Front Immunol 2020; 11:1633. [PMID: 32849557 PMCID: PMC7399169 DOI: 10.3389/fimmu.2020.01633] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 06/18/2020] [Indexed: 12/16/2022] Open
Abstract
The clinical success of cancer immunotherapies targeting PD-1 and CTLA-4 has ignited a substantial research effort to improve our understanding of tumor immunity. Recent studies have revealed that the immune contexture of a tumor influences therapeutic response and survival benefit for cancer patients. Identifying treatment modalities that limit immunosuppression, relieve T cell exhaustion, and potentiate effector functions in the tumor microenvironment (TME) is of much interest. In particular, combinatorial therapeutic approaches that re-educate the TME by limiting the accumulation of immunosuppressive immune cells, such as Foxp3 regulatory T cells (Tregs) and tumor-associated macrophages (TAMs), while promoting CD8+ and CD4+ effector T cell activity is critical. Here, we review key approaches to target these immunosuppressive immune cell subsets and signaling molecules and define the impact of these changes to the tumor milieu. We will highlight the preclinical and clinical evidence for their ability to improve anti-tumor immune responses as well as strategies and challenges for their implementation. Together, this review will provide understanding of therapeutic approaches to efficiently shape the TME and reinvigorate the immune response against cancer.
Collapse
Affiliation(s)
- Shin Foong Ngiow
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA, United States
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Immunology, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Arabella Young
- Department of Immunology, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
- Diabetes Center, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
82
|
Saxena P, Singh PK, Malik PS, Singh N. Immunotherapy Alone or in Combination with Chemotherapy as First-Line Treatment of Non-Small Cell Lung Cancer. Curr Treat Options Oncol 2020; 21:69. [PMID: 32720019 DOI: 10.1007/s11864-020-00768-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OPINION STATEMENT Immune checkpoint inhibitors (ICIs) have revolutionized the management of metastatic and selected cases of unresectable advanced non-small cell lung cancer (NSCLC). Importantly for patients, this implies that in the absence of a targetable oncogenic driver [especially epidermal growth factor receptor (EGFR) gene mutations and anaplastic lymphoma kinase (ALK) gene rearrangements] and in the presence of high programmed death-ligand 1 (PD-L1) expression (≥ 50%), they are eligible for mono-therapy with pembrolizumab thereby avoiding chemotherapy as the first line of treatment. This mono-immunotherapy approach for high PD-L1 metastatic NSCLC is associated with improved overall survival (OS) and radiological responses (RR) with lesser toxicity as compared with conventional platinum doublet chemotherapy for both non-squamous and squamous histological types.However, majority of NSCLC patients either have no or low expression of PD-L1 (< 50%) and such patients derive greater benefit from a combination of PD-1/PD-L1 ICIs with platinum doublet chemotherapy as compared with chemotherapy alone. Again, benefits are seen for both OS and RRs. However, combining immunotherapy with chemotherapy, in general, does lead to higher toxicity than those seen with either of the two alone.Additionally, for non-squamous NSCLC patients, clinicians should not initiate ICI treatment till the results of common targetable genetic alterations like EGFR mutation, ALK, and ROS1 gene rearrangement testing are known (preferably on broad next generation sequencing) and are negative (even if results of PD-L1 testing are available)-as targeted therapies remain the cornerstone of treatment for patients harboring these oncogenic drivers.It is worth mentioning that PD-1 and PD-L1 ICIs are very expensive, and their usage is associated with occurrence of immune-related adverse events (irAEs) which occasionally can be severe. Hence, it is important to discuss efficacy, toxicity, and cost-related to ICI treatment with each and every patient at diagnosis in order to help them decide if they are willing to go ahead with this form of therapy either singly (for high PD-L1 expressors) or in combination with chemotherapy (for others).
Collapse
Affiliation(s)
- Puneet Saxena
- Department of Pulmonary Medicine, Postgraduate Institute of Medical Education and Research (PGIMER), Sector 12, Chandigarh, 160012, India
| | - Pawan Kumar Singh
- Department of Pulmonary and Critical Care Medicine, Post Graduate Institute of Medical Sciences (PGIMS), University of Health Sciences, Rohtak, Haryana, India
| | - Prabhat Singh Malik
- Department of Medical Oncology, Dr. B.R.A.I.R.C.H, All India Institute of Medical Sciences, New Delhi, India
| | - Navneet Singh
- Department of Pulmonary Medicine, Postgraduate Institute of Medical Education and Research (PGIMER), Sector 12, Chandigarh, 160012, India.
| |
Collapse
|
83
|
Ciner AT, Jones K, Muschel RJ, Brodt P. The unique immune microenvironment of liver metastases: Challenges and opportunities. Semin Cancer Biol 2020; 71:143-156. [PMID: 32526354 DOI: 10.1016/j.semcancer.2020.06.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 06/04/2020] [Accepted: 06/05/2020] [Indexed: 02/07/2023]
Abstract
Liver metastases from gastrointestinal and non-gastrointestinal malignancies remain a major cause of cancer-related mortality and a major clinical challenge. The liver has unique properties that facilitate metastatic expansion, including a complex immune system that evolved to dampen immunity to neoantigens entering the liver from the gut, through the portal circulation. In this review, we describe the unique microenvironment encountered by cancer cells in the liver, focusing on elements of the innate and adaptive immune response that can act as a double-edge sword, contributing to the elimination of cancer cells on the one hand and promoting their survival and growth, on the other. We discuss this microenvironment in a clinical context, particularly for colorectal carcinoma, and highlight how a better understanding of the role of the microenvironment has spurred an intense effort to develop novel and innovative strategies for targeting liver metastatic disease, some of which are currently being tested in the clinic.
Collapse
Affiliation(s)
- Aaron T Ciner
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
| | - Keaton Jones
- Oxford Institute for Radiation Oncology, Department of Surgery, University of Oxford, Oxford, UK
| | - Ruth J Muschel
- Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, UK
| | - Pnina Brodt
- Departments of Surgery, Medicine and Oncology, McGill University, and the Research Institute of the McGill University Health Center, Montreal, QC, Canada.
| |
Collapse
|
84
|
Wischhusen J, Melero I, Fridman WH. Growth/Differentiation Factor-15 (GDF-15): From Biomarker to Novel Targetable Immune Checkpoint. Front Immunol 2020; 11:951. [PMID: 32508832 PMCID: PMC7248355 DOI: 10.3389/fimmu.2020.00951] [Citation(s) in RCA: 269] [Impact Index Per Article: 53.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 04/23/2020] [Indexed: 12/12/2022] Open
Abstract
Growth/differentiation factor-15 (GDF-15), also named macrophage inhibitory cytokine-1, is a divergent member of the transforming growth factor β superfamily. While physiological expression is barely detectable in most somatic tissues in humans, GDF-15 is abundant in placenta. Elsewhere, GDF-15 is often induced under stress conditions, seemingly to maintain cell and tissue homeostasis; however, a moderate increase in GDF-15 blood levels is observed with age. Highly elevated GDF-15 levels are mostly linked to pathological conditions including inflammation, myocardial ischemia, and notably cancer. GDF-15 has thus been widely explored as a biomarker for disease prognosis. Mechanistically, induction of anorexia via the brainstem-restricted GDF-15 receptor GFRAL (glial cell-derived neurotrophic factor [GDNF] family receptor α-like) is well-documented. GDF-15 and GFRAL have thus become attractive targets for metabolic intervention. Still, several GDF-15 mediated effects (including its physiological role in pregnancy) are difficult to explain via the described pathway. Hence, there is a clear need to better understand non-metabolic effects of GDF-15. With particular emphasis on its immunomodulatory potential this review discusses the roles of GDF-15 in pregnancy and in pathological conditions including myocardial infarction, autoimmune disease, and specifically cancer. Importantly, the strong predictive value of GDF-15 as biomarker may plausibly be linked to its immune-regulatory function. The described associations and mechanistic data support the hypothesis that GDF-15 acts as immune checkpoint and is thus an emerging target for cancer immunotherapy.
Collapse
Affiliation(s)
- Jörg Wischhusen
- Experimental Tumor Immunology, Department of Obstetrics and Gynecology, University of Würzburg Medical School, Würzburg, Germany
| | - Ignacio Melero
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
- Centro de Investigación Biomédica en Red Cáncer, CIBERONC, Madrid, Spain
- Immunology and Immunotherapy Unit, Clínica Universidad de Navarra, Pamplona, Spain
| | - Wolf Herman Fridman
- INSERM, UMR_S 1138, Cordeliers Research Center, Université de Paris, Sorbonne Université Team Cancer, Immune Control and Escape, Paris, France
| |
Collapse
|
85
|
Schullian P, Putzer D, Eberle G, Laimer G, Bale R. Simultaneous Stereotactic Radiofrequency Ablation of Multiple (≥ 4) Liver Tumors: Feasibility, Safety, and Efficacy. J Vasc Interv Radiol 2020; 31:943-952. [PMID: 32414570 DOI: 10.1016/j.jvir.2019.12.794] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 12/15/2019] [Accepted: 12/23/2019] [Indexed: 12/19/2022] Open
Abstract
PURPOSE To assess feasibility, safety, and clinical outcome of simultaneous stereotactic radiofrequency (RF) ablation of multiple (≥ 4) primary and metastatic liver tumors. MATERIALS AND METHODS This retrospective observational study included 92 patients (29 women, 62 men), 35 with ≥ 4 hepatocellular carcinomas (HCCs) and 57 with ≥ 4 metastatic liver tumors at initial stereotactic RF ablation between 2005 and 2018. The median size of 178 HCCs and 371 metastases was 2.2 cm (range, 1.0-8.5 cm) and 3.0 cm (range, 0.5-13 cm), respectively. At initial stereotactic RF ablation, 17 (48.6%) patients with HCC and 19 (33.3%) with metastases had 4 liver tumors, 11 (31.4%) and 19 (33.3%) had 5 tumors, and 7 (20%) and 19 (33.3%) had ≥ 6 tumors. RESULTS Major complications occurred in 2 of 35 ablations (5.4%) in patients with HCCs and in 7 of 63 (10%) with metastases. The primary technical efficacy rate (ie, successful initial ablation) was 100% (178/178) in HCCs and 98.8% (363/371) in metastases. Local recurrence was observed in 4 of 178 (2.2%) HCCs and in 17 of 371 (4.6%) metastases. Overall survival (OS) rates at 1, 3, and 5 years from the date of the first stereotactic RF ablation were 88.0%, 54.0%, and 30.4% for patients with HCCs with a median OS of 38.2 months and 86.1%, 53.1%, and 37.3% for patients with metastases with a median OS of 37.4 months. CONCLUSIONS Stereotactic RF ablation is a feasible, safe, and efficacious option in simultaneous management of multiple primary and metastatic liver tumors.
Collapse
Affiliation(s)
- Peter Schullian
- Section of Interventional Oncology-Microinvasive Therapy (SIP), Department of Radiology, Medical University of Innsbruck, Anichstrasse 35, Innsbruck 6020, Austria
| | - Daniel Putzer
- Section of Interventional Oncology-Microinvasive Therapy (SIP), Department of Radiology, Medical University of Innsbruck, Anichstrasse 35, Innsbruck 6020, Austria
| | - Gernot Eberle
- Section of Interventional Oncology-Microinvasive Therapy (SIP), Department of Radiology, Medical University of Innsbruck, Anichstrasse 35, Innsbruck 6020, Austria
| | - Gregor Laimer
- Section of Interventional Oncology-Microinvasive Therapy (SIP), Department of Radiology, Medical University of Innsbruck, Anichstrasse 35, Innsbruck 6020, Austria
| | - Reto Bale
- Section of Interventional Oncology-Microinvasive Therapy (SIP), Department of Radiology, Medical University of Innsbruck, Anichstrasse 35, Innsbruck 6020, Austria.
| |
Collapse
|
86
|
Botticelli A, Cirillo A, Scagnoli S, Cerbelli B, Strigari L, Cortellini A, Pizzuti L, Vici P, De Galitiis F, Di Pietro FR, Cerbelli E, Ghidini M, D’Amati G, Della Rocca C, Mezi S, Gelibter A, Giusti R, Cortesi E, Ascierto PA, Nuti M, Marchetti P. The Agnostic Role of Site of Metastasis in Predicting Outcomes in Cancer Patients Treated with Immunotherapy. Vaccines (Basel) 2020; 8:vaccines8020203. [PMID: 32353934 PMCID: PMC7349154 DOI: 10.3390/vaccines8020203] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 04/23/2020] [Accepted: 04/25/2020] [Indexed: 12/19/2022] Open
Abstract
Immune checkpoint inhibitors have revolutionized treatment and outcome of melanoma and many other solid malignancies including non-small cell lung cancer (NSCLC) and renal cell carcinoma (RCC). Unfortunately, only a minority of patients have a long-term benefit, while the remaining demonstrate primary or acquired resistance. Recently, it has been demonstrated that the prevalence of programmed death-ligand 1 (PD-L1) and tumor-infiltrating lymphocytes (TILs) varies based on the anatomical site of metastases. In particular, liver seems to have more immunosuppressive microenvironment while both the presence of lymph nodal disease and lung metastases seem to have the highest prevalence of PD-L1 and TILs. The aim of the present study is to investigate the possible role of site of metastases as a predictive factor for response or resistance to immunotherapy in several types of cancer. In this multicenter retrospective study, we enrolled patients with metastatic NSCLC, melanoma, RCC, urothelial, merkel carcinoma, and colon cancer who received immunotherapy from April 2015 to August 2019. Major clinicopathological parameters were retrieved and correlated with patients’ survival outcomes in order to assess their prognostic value and build a useful tool to assist in the decision-making process. A total of 291 patients were included in this study. One hundred eighty-seven (64%) patients were male and 104 (36%) female. The tumor histology was squamous NSCLC in 56 (19%) patients, non-squamous NSCLC in 99 (34%) patients, melanoma in 101 (35%) patients, RCC in 28 (10%) patients, and other tumors in the remaining 7 (2%) patients. The number of metastatic sites was 1 in 103 patients (35%), 2 in 104 patients (36%) and 3 in 84 patients (29%). Out of 183 valuable patients, the entity of response was complete response (CR), partial response (PR), stable disease (SD), and progression disease (PD) in 15, 53, 31, and 79 patients, respectively. Using an univariate analysis (UVA), tumor burden (p = 0.0004), the presence of liver (p = 0.0009), bone (p = 0.0016), brain metastases (p < 0.0001), the other metastatic sites (p = 0.0375), the number of metastatic sites (p = 0.0039), the histology (p = 0.0034), the upfront use of immunotherapy (p = 0.0032), and Eastern Cooperative Oncology Group (ECOG) Perfomance status (PS) ≥ 1 (p < 0.0001) were significantly associated with poor overall survival (OS). Using a multivariate analysis (MVA) the presence of liver (p = 0.0105) and brain (p = 0.0026) metastases, the NSCLC diagnosis (p < 0.0001) and the ECOG PS (p < 0.0001) resulted as significant prognostic factors of survival. Regarding the progression free survival (PFS), using a UVA of the tumor burden (p = 0.0004), bone (p = 0.0098) and brain (p = 0.0038) metastases, the presence of other metastatic sites (p = 0.0063), the number of metastatic sites (p = 0.0007), the histology (p = 0.0007), the use of immunotherapy as first line (p = 0.0031), and the ECOG PS ≥ 1 (p ≤ 0.0001) were associated with a lower PFS rate. Using an MVA, the presence of brain (p = 0.0088) and liver metastases (p = 0.024) and the ECOG PS (p < 0.0001) resulted as predictors of poor PFS. Our study suggests that the site of metastases could have a role as prognostic and predictive factor in patients treated with immunotherapy. Indeed, regardless of the histology, the presence of liver and brain metastases was associated with a shorter PFS and OS, but these results must be confirmed in further studies. In this context, a deep characterization of microenvironment could be crucial to prepare patients through novel strategies with combination or sequential immunotherapy in order to improve treatment response.
Collapse
Affiliation(s)
- Andrea Botticelli
- Department of Clinical and molecular oncology, University of Rome “Sapienza”, 00185 Rome, Italy; (A.B.); (P.M.)
| | - Alessio Cirillo
- Department of Radiological, Oncological and anatomo-pathological Science, University of Rome “Sapienza”, 00185 Rome, Italy; (A.C.); (B.C.); (E.C.); (G.D.); (S.M.); (A.G.); (E.C.)
| | - Simone Scagnoli
- Department of Medical and Surgical Sciences and Translational Medicine, University of Rome “Sapienza”, 00185 Rome, Italy
- Correspondence: ; Tel.: +39-3280306525
| | - Bruna Cerbelli
- Department of Radiological, Oncological and anatomo-pathological Science, University of Rome “Sapienza”, 00185 Rome, Italy; (A.C.); (B.C.); (E.C.); (G.D.); (S.M.); (A.G.); (E.C.)
| | - Lidia Strigari
- Medical Physics Unit, “S. Orsola-Malpighi” Hospital, 40138 Bologna, Italy;
| | - Alessio Cortellini
- Department of Biotechnology and Applied Clinical Sciences, University of L’Aquila, 671000 L’Aquila, Italy; (A.C.); (L.P.)
| | - Laura Pizzuti
- Department of Biotechnology and Applied Clinical Sciences, University of L’Aquila, 671000 L’Aquila, Italy; (A.C.); (L.P.)
| | - Patrizia Vici
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy;
| | | | | | - Edoardo Cerbelli
- Department of Radiological, Oncological and anatomo-pathological Science, University of Rome “Sapienza”, 00185 Rome, Italy; (A.C.); (B.C.); (E.C.); (G.D.); (S.M.); (A.G.); (E.C.)
| | - Michele Ghidini
- Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy;
| | - Giulia D’Amati
- Department of Radiological, Oncological and anatomo-pathological Science, University of Rome “Sapienza”, 00185 Rome, Italy; (A.C.); (B.C.); (E.C.); (G.D.); (S.M.); (A.G.); (E.C.)
| | - Carlo Della Rocca
- Department of Medico-Surgical Sciences and Biotechnology, Polo Pontino, Sapienza University, 00185 Roma, Italy;
| | - Silvia Mezi
- Department of Radiological, Oncological and anatomo-pathological Science, University of Rome “Sapienza”, 00185 Rome, Italy; (A.C.); (B.C.); (E.C.); (G.D.); (S.M.); (A.G.); (E.C.)
| | - Alain Gelibter
- Department of Radiological, Oncological and anatomo-pathological Science, University of Rome “Sapienza”, 00185 Rome, Italy; (A.C.); (B.C.); (E.C.); (G.D.); (S.M.); (A.G.); (E.C.)
| | - Raffaele Giusti
- Sant’Andrea Hospital, Department of clinical and molecular medicine, Sapienza, University of Rome, 00153 Roma, Italy;
| | - Enrico Cortesi
- Department of Radiological, Oncological and anatomo-pathological Science, University of Rome “Sapienza”, 00185 Rome, Italy; (A.C.); (B.C.); (E.C.); (G.D.); (S.M.); (A.G.); (E.C.)
| | - Paolo Antonio Ascierto
- Unit of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Via Mariano Semmola snc, 80131 Naples, Italy;
| | - Marianna Nuti
- Department of Experimental Medicine, University Sapienza, 00185 Rome, Italy;
| | - Paolo Marchetti
- Department of Clinical and molecular oncology, University of Rome “Sapienza”, 00185 Rome, Italy; (A.B.); (P.M.)
| |
Collapse
|
87
|
Amaral T, Seeber O, Mersi E, Sanchez S, Thomas I, Meiwes A, Forschner A, Leiter U, Eigentler T, Keim U, Garbe C. Primary Resistance to PD-1-Based Immunotherapy-A Study in 319 Patients with Stage IV Melanoma. Cancers (Basel) 2020; 12:cancers12041027. [PMID: 32331243 PMCID: PMC7226601 DOI: 10.3390/cancers12041027] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 04/16/2020] [Accepted: 04/20/2020] [Indexed: 02/06/2023] Open
Abstract
Background: Primary resistance to immunotherapy can be observed in approximately 40–65% of the stage IV melanoma patients treated with immune checkpoint inhibitors. A minority of the patients receive a second-line therapy, and the clinical benefit is small. Patients and methods: Stage IV melanoma patients treated with first-line PD-1-based immunotherapy between January 2015 and December 2018 were investigated. Primary resistance was defined as progressive disease (PD) at the time of the first tumor assessment after starting immunotherapy. Patients with complete response, partial response, and stable disease were classified as having disease control (DC). Overall survival (OS) and progression-free survival (PFS) were evaluated by Kaplan–Meier estimator. Univariate and multivariate logistic regression analyses were performed to determine prognostic factors associated with OS. Results: Three hundred and nineteen patients were included, and 40% had primary resistance to immunotherapy. The median follow-up time was 22 months. Patients with primary resistance had 1-, 2-, and 3-year OS rates of 41%, 15%, and 10%, respectively, compared to 91%, 81%, and 65% for the patients who achieved DC. The following independently significant prognostic factors for OS were identified: protein S100B level and primary tumor localization. There was a statistically significant difference for OS (p < 0.0001) but not for PFS (p = 0.230) when analyzing risk groups formed with a combination of these two variables (low-, intermediate-, and high-risk subgroups). Conclusions: Melanoma patients with primary resistance to immunotherapy have a dismal prognosis. Response at the first tumor assessment after starting immunotherapy is a stronger prognostic factor for the further course of the disease than pretreatment risk factors.
Collapse
Affiliation(s)
- Teresa Amaral
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University of Tuebingen, 72076 Tuebingen, Germany; (T.A.)
- Portuguese Air Force—Health Care Direction, 1649-020 Lisbon, Portugal
| | - Olivia Seeber
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University of Tuebingen, 72076 Tuebingen, Germany; (T.A.)
| | - Edgar Mersi
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University of Tuebingen, 72076 Tuebingen, Germany; (T.A.)
| | - Stephanie Sanchez
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University of Tuebingen, 72076 Tuebingen, Germany; (T.A.)
| | - Ioannis Thomas
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University of Tuebingen, 72076 Tuebingen, Germany; (T.A.)
| | - Andreas Meiwes
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University of Tuebingen, 72076 Tuebingen, Germany; (T.A.)
| | - Andrea Forschner
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University of Tuebingen, 72076 Tuebingen, Germany; (T.A.)
| | - Ulrike Leiter
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University of Tuebingen, 72076 Tuebingen, Germany; (T.A.)
| | - Thomas Eigentler
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University of Tuebingen, 72076 Tuebingen, Germany; (T.A.)
| | - Ulrike Keim
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University of Tuebingen, 72076 Tuebingen, Germany; (T.A.)
| | - Claus Garbe
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University of Tuebingen, 72076 Tuebingen, Germany; (T.A.)
- Correspondence: ; Tel.: +49-7071-298-87110; Fax: +49-7071-29-51-87
| |
Collapse
|
88
|
Bianchi JJ, Zhao X, Mays JC, Davoli T. Not all cancers are created equal: Tissue specificity in cancer genes and pathways. Curr Opin Cell Biol 2020; 63:135-143. [PMID: 32092639 PMCID: PMC7247947 DOI: 10.1016/j.ceb.2020.01.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 01/02/2020] [Accepted: 01/07/2020] [Indexed: 12/22/2022]
Abstract
Tumors arise through waves of genetic alterations and clonal expansion that allow tumor cells to acquire cancer hallmarks, such as genome instability and immune evasion. Recent genomic analyses showed that the vast majority of cancer driver genes are mutated in a tissue-dependent manner, that is, are altered in some cancers but not others. Often the tumor type also affects the likelihood of therapy response. What is the origin of tissue specificity in cancer? Recent studies suggest that both cell-intrinsic and cell-extrinsic factors play a role. On one hand, cell type-specific wiring of the cell signaling network determines the outcome of cancer driver gene mutations. On the other hand, the tumor cells' exposure to tissue-specific microenvironments (e.g. immune cells) also contributes to shape the tissue specificity of driver genes and of therapy response. In the future, a more complete understanding of tissue specificity in cancer may inform methods to better predict and improve therapeutic outcomes.
Collapse
Affiliation(s)
- Joy J Bianchi
- Institute for Systems Genetics, NYU Langone Health, New York, NY 10016, USA; Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, 10016, USA
| | - Xin Zhao
- Institute for Systems Genetics, NYU Langone Health, New York, NY 10016, USA; Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, 10016, USA
| | - Joseph C Mays
- Institute for Systems Genetics, NYU Langone Health, New York, NY 10016, USA; Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, 10016, USA
| | - Teresa Davoli
- Institute for Systems Genetics, NYU Langone Health, New York, NY 10016, USA; Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, 10016, USA.
| |
Collapse
|
89
|
Köstek O, Demir T. Sarcopenia and Inflammation with Immunotherapy. Oncologist 2020; 25:e875. [PMID: 32191391 DOI: 10.1634/theoncologist.2019-1005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 02/12/2020] [Indexed: 11/17/2022] Open
Affiliation(s)
- Osman Köstek
- Edirne Sultan I. Murat State Hospital, Clinic of Medical Oncology, Edirne, Turkey
| | - Tarık Demir
- Haydarpaşa Numune Training and Research Hospital, Clinic of Medical Oncology, İstanbul, Turkey
| |
Collapse
|
90
|
Shabto JM, Martini DJ, Liu Y, Ravindranathan D, Brown J, Hitron EE, Russler GA, Caulfield S, Kissick H, Alemozaffar M, Ogan K, Harris WB, Master VA, Kucuk O, Carthon BC, Bilen MA. Novel risk group stratification for metastatic urothelial cancer patients treated with immune checkpoint inhibitors. Cancer Med 2020; 9:2752-2760. [PMID: 32100417 PMCID: PMC7163104 DOI: 10.1002/cam4.2932] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 02/01/2020] [Accepted: 02/03/2020] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND We developed a novel risk scoring system for urothelial cancer (UC) patients receiving immune checkpoint inhibitors (ICI). METHODS We conducted a retrospective review of 67 UC patients treated with ICI at Winship Cancer Institute of Emory University from 2015 to 2018. Using stepwise variable selection in Cox proportional hazard model and Sullivan's weighting schema, baseline platelet-to-lymphocyte ratio (PLR), presence of liver metastasis, baseline albumin, and baseline Eastern Cooperative Oncology Group performance status (ECOG PS) were used for risk scoring. Patients were categorized into good risk (risk score 0-1), intermediate risk (risk score 2-3), and poor risk (risk score 4-6). Univariable (UVA) and multivariable analysis (MVA) and Kaplan-Meier method were used to assess overall survival (OS) and progression free survival (PFS). RESULTS The Emory Risk Scoring System had C-statistics of 0.74 (Standard Error = 0.047) in predicting OS and 0.70 (Standard Error = 0.043) in predicting PFS. Compared to good risk patients, poor risk patients had significantly shorter OS and PFS in both UVA and MVA (all P < .001), and intermediate risk patients had significantly shorter OS and PFS in both UVA and MVA (all P < .03). CONCLUSIONS Risk scoring using baseline PLR, presence of liver metastasis, baseline albumin, and baseline ECOG PS may effectively predict OS and PFS in UC patients receiving ICI.
Collapse
Affiliation(s)
- Julie M Shabto
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA.,Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Dylan J Martini
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA.,Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Yuan Liu
- Departments of Biostatistics and Bioinformatics, Emory University, Atlanta, GA, USA
| | - Deepak Ravindranathan
- Winship Cancer Institute of Emory University, Atlanta, GA, USA.,Department of Medicine, Emory University, Atlanta, GA, USA
| | - Jacqueline Brown
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA.,Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Emilie E Hitron
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Greta A Russler
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA.,Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Sarah Caulfield
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA.,Department of Pharmaceutical Services, Emory University School of Medicine, Atlanta, GA, USA
| | - Haydn Kissick
- Winship Cancer Institute of Emory University, Atlanta, GA, USA.,Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
| | - Mehrdad Alemozaffar
- Winship Cancer Institute of Emory University, Atlanta, GA, USA.,Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
| | - Kenneth Ogan
- Winship Cancer Institute of Emory University, Atlanta, GA, USA.,Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
| | - Wayne B Harris
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA.,Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Viraj A Master
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
| | - Omer Kucuk
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA.,Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Bradley C Carthon
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA.,Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Mehmet A Bilen
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA.,Winship Cancer Institute of Emory University, Atlanta, GA, USA
| |
Collapse
|
91
|
Prelaj A, Ferrara R, Rebuzzi SE, Proto C, Signorelli D, Galli G, De Toma A, Randon G, Pagani F, Viscardi G, Brambilla M, Trevisan B, Ganzinelli M, Martinetti A, Gallucci R, Di Mauro RM, Molino G, Zilembo N, Torri V, de Braud FM, Garassino MC, Lo Russo G. EPSILoN: A Prognostic Score for Immunotherapy in Advanced Non-Small-Cell Lung Cancer: A Validation Cohort. Cancers (Basel) 2019; 11:E1954. [PMID: 31817541 PMCID: PMC6966664 DOI: 10.3390/cancers11121954] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 12/01/2019] [Accepted: 12/02/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Beyond programmed death ligand 1 (PD-L1), no other biomarkers for immunotherapy are used in daily practice. We previously created EPSILoN (Eastern Cooperative Oncology Group performance status (ECOG PS), smoking, liver metastases, lactate dehydrogenase (LDH), neutrophil-to-lymphocyte ratio (NLR)) score, a clinical/biochemical prognostic score, in 154 patients treated with second/further-line immunotherapy. This study's aim was to validate EPSILoN score in a different population group. METHODS 193 patients were included at National Cancer Institute of Milan (second-line immunotherapy, 61%; further-line immunotherapy, 39%). Clinical/laboratory parameters such as neutrophil-to-lymphocyte ratio and lactate dehydrogenase levels were collected. Kaplan-Meier and Cox hazard methods were used for survival analysis. RESULTS Overall median progression-free survival and median overall survival were 2.3 and 7.6 months, respectively. Multivariate analyses for Progression-Free Survival (PFS) identified heavy smokers (hazard ratio (HR) 0.71, p = 0.036) and baseline LDH < 400 mg/dL (HR 0.66, p = 0.026) as independent positive factors and liver metastases (HR 1.48, p = 0.04) and NLR ≥ 4 (HR 1.49, p = 0.029) as negative prognostic factors. These five factors were included in the EPSILoN score which was able to stratify patients in three different prognostic groups, high, intermediate and low, with PFS of 6.0, 3.8 and 1.9 months, respectively (HR 1.94, p < 0.001); high, intermediate and low prognostic groups had overall survival (OS) of 24.5, 8.9 and 3.4 months, respectively (HR 2.40, p < 0.001). CONCLUSIONS EPSILoN, combining five baseline clinical/blood parameters (ECOG PS, smoking, liver metastases, LDH, NLR), may help to identify advanced non-small-cell lung cancer (aNSCLC) patients who most likely benefit from immune checkpoint inhibitors (ICIs).
Collapse
Affiliation(s)
- Arsela Prelaj
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, 20133 Milan, Italy; (R.F.); (C.P.); (D.S.); (G.G.); (A.D.T.); (G.R.); (F.P.); (G.V.); (M.B.); (B.T.); (M.G.); (A.M.); (R.G.); (R.M.D.M.); (G.M.); (N.Z.); (F.M.d.B.); (M.C.G.); (G.L.R.)
| | - Roberto Ferrara
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, 20133 Milan, Italy; (R.F.); (C.P.); (D.S.); (G.G.); (A.D.T.); (G.R.); (F.P.); (G.V.); (M.B.); (B.T.); (M.G.); (A.M.); (R.G.); (R.M.D.M.); (G.M.); (N.Z.); (F.M.d.B.); (M.C.G.); (G.L.R.)
| | - Sara Elena Rebuzzi
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy;
| | - Claudia Proto
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, 20133 Milan, Italy; (R.F.); (C.P.); (D.S.); (G.G.); (A.D.T.); (G.R.); (F.P.); (G.V.); (M.B.); (B.T.); (M.G.); (A.M.); (R.G.); (R.M.D.M.); (G.M.); (N.Z.); (F.M.d.B.); (M.C.G.); (G.L.R.)
| | - Diego Signorelli
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, 20133 Milan, Italy; (R.F.); (C.P.); (D.S.); (G.G.); (A.D.T.); (G.R.); (F.P.); (G.V.); (M.B.); (B.T.); (M.G.); (A.M.); (R.G.); (R.M.D.M.); (G.M.); (N.Z.); (F.M.d.B.); (M.C.G.); (G.L.R.)
| | - Giulia Galli
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, 20133 Milan, Italy; (R.F.); (C.P.); (D.S.); (G.G.); (A.D.T.); (G.R.); (F.P.); (G.V.); (M.B.); (B.T.); (M.G.); (A.M.); (R.G.); (R.M.D.M.); (G.M.); (N.Z.); (F.M.d.B.); (M.C.G.); (G.L.R.)
| | - Alessandro De Toma
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, 20133 Milan, Italy; (R.F.); (C.P.); (D.S.); (G.G.); (A.D.T.); (G.R.); (F.P.); (G.V.); (M.B.); (B.T.); (M.G.); (A.M.); (R.G.); (R.M.D.M.); (G.M.); (N.Z.); (F.M.d.B.); (M.C.G.); (G.L.R.)
| | - Giovanni Randon
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, 20133 Milan, Italy; (R.F.); (C.P.); (D.S.); (G.G.); (A.D.T.); (G.R.); (F.P.); (G.V.); (M.B.); (B.T.); (M.G.); (A.M.); (R.G.); (R.M.D.M.); (G.M.); (N.Z.); (F.M.d.B.); (M.C.G.); (G.L.R.)
| | - Filippo Pagani
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, 20133 Milan, Italy; (R.F.); (C.P.); (D.S.); (G.G.); (A.D.T.); (G.R.); (F.P.); (G.V.); (M.B.); (B.T.); (M.G.); (A.M.); (R.G.); (R.M.D.M.); (G.M.); (N.Z.); (F.M.d.B.); (M.C.G.); (G.L.R.)
| | - Giuseppe Viscardi
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, 20133 Milan, Italy; (R.F.); (C.P.); (D.S.); (G.G.); (A.D.T.); (G.R.); (F.P.); (G.V.); (M.B.); (B.T.); (M.G.); (A.M.); (R.G.); (R.M.D.M.); (G.M.); (N.Z.); (F.M.d.B.); (M.C.G.); (G.L.R.)
| | - Marta Brambilla
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, 20133 Milan, Italy; (R.F.); (C.P.); (D.S.); (G.G.); (A.D.T.); (G.R.); (F.P.); (G.V.); (M.B.); (B.T.); (M.G.); (A.M.); (R.G.); (R.M.D.M.); (G.M.); (N.Z.); (F.M.d.B.); (M.C.G.); (G.L.R.)
| | - Benedetta Trevisan
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, 20133 Milan, Italy; (R.F.); (C.P.); (D.S.); (G.G.); (A.D.T.); (G.R.); (F.P.); (G.V.); (M.B.); (B.T.); (M.G.); (A.M.); (R.G.); (R.M.D.M.); (G.M.); (N.Z.); (F.M.d.B.); (M.C.G.); (G.L.R.)
| | - Monica Ganzinelli
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, 20133 Milan, Italy; (R.F.); (C.P.); (D.S.); (G.G.); (A.D.T.); (G.R.); (F.P.); (G.V.); (M.B.); (B.T.); (M.G.); (A.M.); (R.G.); (R.M.D.M.); (G.M.); (N.Z.); (F.M.d.B.); (M.C.G.); (G.L.R.)
| | - Antonia Martinetti
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, 20133 Milan, Italy; (R.F.); (C.P.); (D.S.); (G.G.); (A.D.T.); (G.R.); (F.P.); (G.V.); (M.B.); (B.T.); (M.G.); (A.M.); (R.G.); (R.M.D.M.); (G.M.); (N.Z.); (F.M.d.B.); (M.C.G.); (G.L.R.)
| | - Rosaria Gallucci
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, 20133 Milan, Italy; (R.F.); (C.P.); (D.S.); (G.G.); (A.D.T.); (G.R.); (F.P.); (G.V.); (M.B.); (B.T.); (M.G.); (A.M.); (R.G.); (R.M.D.M.); (G.M.); (N.Z.); (F.M.d.B.); (M.C.G.); (G.L.R.)
| | - Rosa Maria Di Mauro
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, 20133 Milan, Italy; (R.F.); (C.P.); (D.S.); (G.G.); (A.D.T.); (G.R.); (F.P.); (G.V.); (M.B.); (B.T.); (M.G.); (A.M.); (R.G.); (R.M.D.M.); (G.M.); (N.Z.); (F.M.d.B.); (M.C.G.); (G.L.R.)
| | - Giuliano Molino
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, 20133 Milan, Italy; (R.F.); (C.P.); (D.S.); (G.G.); (A.D.T.); (G.R.); (F.P.); (G.V.); (M.B.); (B.T.); (M.G.); (A.M.); (R.G.); (R.M.D.M.); (G.M.); (N.Z.); (F.M.d.B.); (M.C.G.); (G.L.R.)
| | - Nicoletta Zilembo
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, 20133 Milan, Italy; (R.F.); (C.P.); (D.S.); (G.G.); (A.D.T.); (G.R.); (F.P.); (G.V.); (M.B.); (B.T.); (M.G.); (A.M.); (R.G.); (R.M.D.M.); (G.M.); (N.Z.); (F.M.d.B.); (M.C.G.); (G.L.R.)
| | - Valter Torri
- Pharmacological Research Institute IRCSS Mario Negri, Via La Masa 19, 20156 Milan, Italy;
| | - Filippo Maria de Braud
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, 20133 Milan, Italy; (R.F.); (C.P.); (D.S.); (G.G.); (A.D.T.); (G.R.); (F.P.); (G.V.); (M.B.); (B.T.); (M.G.); (A.M.); (R.G.); (R.M.D.M.); (G.M.); (N.Z.); (F.M.d.B.); (M.C.G.); (G.L.R.)
| | - Marina Chiara Garassino
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, 20133 Milan, Italy; (R.F.); (C.P.); (D.S.); (G.G.); (A.D.T.); (G.R.); (F.P.); (G.V.); (M.B.); (B.T.); (M.G.); (A.M.); (R.G.); (R.M.D.M.); (G.M.); (N.Z.); (F.M.d.B.); (M.C.G.); (G.L.R.)
| | - Giuseppe Lo Russo
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, 20133 Milan, Italy; (R.F.); (C.P.); (D.S.); (G.G.); (A.D.T.); (G.R.); (F.P.); (G.V.); (M.B.); (B.T.); (M.G.); (A.M.); (R.G.); (R.M.D.M.); (G.M.); (N.Z.); (F.M.d.B.); (M.C.G.); (G.L.R.)
| |
Collapse
|
92
|
De Giglio A, Nuvola G, Dall'Olio FG. Carboplatin, nab-paclitaxel plus atezolizumab in IMpower 130 trial: new weapons beyond controversies. Transl Cancer Res 2019; 8:E18-E23. [PMID: 35117058 PMCID: PMC8798162 DOI: 10.21037/tcr.2019.12.69] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 12/19/2019] [Indexed: 11/06/2022]
Affiliation(s)
- Andrea De Giglio
- Department of Specialized, Experimental and Diagnostic Medicine, S.Orsola-Malpighi University Hospital, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Giacomo Nuvola
- Department of Specialized, Experimental and Diagnostic Medicine, S.Orsola-Malpighi University Hospital, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Filippo Gustavo Dall'Olio
- Department of Specialized, Experimental and Diagnostic Medicine, S.Orsola-Malpighi University Hospital, Alma Mater Studiorum University of Bologna, Bologna, Italy
| |
Collapse
|