1
|
Ditonno F, Bologna E, Licari LC, Franco A, Pettenuzzo G, Leonardo C, Proietti F, Carbonara U, Mir MC, Crivellaro S, Veccia A, Bertolo R, Antonelli A, Autorino R. Upfront versus deferred cytoreductive nephrectomy following targeted or immunotherapy: a population-based propensity score-matched analysis of perioperative complications. World J Urol 2024; 42:451. [PMID: 39066794 DOI: 10.1007/s00345-024-05156-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 06/27/2024] [Indexed: 07/30/2024] Open
Abstract
PURPOSE To evaluate the incidence of postoperative complications after cytoreductive nephrectomy (CN) following first-line treatment for metastatic renal cell carcinoma (mRCC), and to compare it with postoperative complications of upfront CN. METHODS For this population-based retrospective study, the PearlDiver Mariner database (PearlDiver Technologies, Colorado Springs, CO), a database of insurance billing records was analyzed. Using relevant ICD-9/10 and CPT codes, patients diagnosed with mRCC between 2011 and 2021, who received first-line systemic molecular therapy (SMT), either tyrosine kinase inhibitors (TKI) or immune-checkpoint inhibitors (ICI), were identified. The selected population was stratified into two cohorts according to the timing of CN (deferred: after SMT vs. upfront: before SMT). Propensity-score matching (PSM) was performed as per baseline patients' characteristics to control for potential confounders between the two cohorts. The primary outcome was to compare 30-day postoperative complications rate between patients undergoing upfront vs. deferred CN. RESULTS After PSM, 162 patients who received upfront CN were compared with 162 patients who underwent deferred CN. The overall rate of 30-day postoperative complications was statistically significantly higher in patients undergoing deferred CN (33.9%), compared to patients treated with upfront CN (21%, p < 0.01). In addition, the rate of both medical (26.5% vs. 14.2%, p < 0.01) and surgical (14.8 vs. 7.4%, p = 0.03) complication rate was statistically significantly higher in deferred vs. upfront CN. Multivariable logistic regression analysis revealed that none of the treatment regimens significantly predicted the occurrence of postoperative complications. CONCLUSION Patients undergoing deferred CN experience a higher rates of overall, medical, and surgical 30-day postoperative complications compared to those receiving upfront surgery. Findings from this study should be interpreted within the limitations of this type of analysis.
Collapse
Affiliation(s)
- Francesco Ditonno
- Department of Urology, Rush University Medical Center, 1725 W. Harrison Street, Suite 970, Chicago, IL, 60612, USA
- Department of Urology, Azienda Ospedaliera Universitaria Integrata Verona, University of Verona, Verona, Italy
| | - Eugenio Bologna
- Department of Urology, Rush University Medical Center, 1725 W. Harrison Street, Suite 970, Chicago, IL, 60612, USA
- Department of Maternal-Child and Urological Sciences, Sapienza University Rome, Policlinico Umberto I Hospital, Rome, Italy
| | - Leslie Claire Licari
- Department of Urology, Rush University Medical Center, 1725 W. Harrison Street, Suite 970, Chicago, IL, 60612, USA
- Department of Maternal-Child and Urological Sciences, Sapienza University Rome, Policlinico Umberto I Hospital, Rome, Italy
| | - Antonio Franco
- Department of Urology, Rush University Medical Center, 1725 W. Harrison Street, Suite 970, Chicago, IL, 60612, USA
- Department of Urology, Sant'Andrea Hospital, Sapienza University, Rome, Italy
| | - Greta Pettenuzzo
- Department of Urology, Azienda Ospedaliera Universitaria Integrata Verona, University of Verona, Verona, Italy
- Department of Urology, University of Illinois at Chicago, Chicago, IL, USA
| | - Costantino Leonardo
- Department of Urology, "Regina Elena" National Cancer Institute, Rome, Italy
| | - Flavia Proietti
- Department of Urology, "Regina Elena" National Cancer Institute, Rome, Italy
| | | | - M Carmen Mir
- Department of Urology, Hospital Universitario La Ribera, Valencia, Spain
| | - Simone Crivellaro
- Department of Urology, University of Illinois at Chicago, Chicago, IL, USA
| | - Alessandro Veccia
- Department of Urology, Azienda Ospedaliera Universitaria Integrata Verona, University of Verona, Verona, Italy
| | - Riccardo Bertolo
- Department of Urology, Azienda Ospedaliera Universitaria Integrata Verona, University of Verona, Verona, Italy
| | - Alessandro Antonelli
- Department of Urology, Azienda Ospedaliera Universitaria Integrata Verona, University of Verona, Verona, Italy
| | - Riccardo Autorino
- Department of Urology, Rush University Medical Center, 1725 W. Harrison Street, Suite 970, Chicago, IL, 60612, USA.
| |
Collapse
|
2
|
Yip W, Ghoreifi A, Gerald T, Lee R, Howard J, Asghar A, Khanna A, Cai J, Aron M, Gill I, Thompson RH, Uzzo R, Margulis V, Singla N, Djaladat H. Perioperative Complications and Oncologic Outcomes of Nephrectomy Following Immune Checkpoint Inhibitor Therapy: A Multicenter Collaborative Study. Eur Urol Oncol 2023; 6:604-610. [PMID: 37005212 DOI: 10.1016/j.euo.2023.03.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 02/14/2023] [Accepted: 03/11/2023] [Indexed: 04/03/2023]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) are now a mainstay of metastatic renal cell carcinoma (RCC) management with five current Food and Drug Administration-approved regimens. However, data regarding nephrectomy outcomes following an ICI are limited. OBJECTIVE To evaluate the safety and outcomes of nephrectomy following an ICI. DESIGN, SETTING, AND PARTICIPANTS A retrospective review was performed of patients with primary locally advanced or metastatic RCC undergoing nephrectomy following an ICI in five US academic centers between January 2011 and September 2021. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Clinical data, perioperative outcomes, and 90-d complications/readmissions were recorded and evaluated by univariate and logistic regression models. Recurrence-free and overall survival probabilities were estimated by the Kaplan-Meier method. RESULTS AND LIMITATIONS A total of 113 patients with a median (interquartile range) age of 63 (56-69) yr were included. The main ICI regimens were nivolumab ± ipilimumab (n = 85) and pembrolizumab ± axitinib (n = 24). Risk groups included 95% intermediate- and 5% poor-risk patients. Surgical procedures were 109 radical and four partial nephrectomies, including 60 open, 38 robotic, and 14 laparoscopic with five (10%) conversions. Two intraoperative complications were reported (bowel and pancreatic injury). The median operative time, estimated blood loss, and hospital stay were 3 h, 250 ml, and 3 d, respectively. A complete pathologic response (ypT0N0) was noted in six (5%) patients. The 90-d complication rate was 24%, with 12 (11%) patients requiring readmission. On a multivariable analysis, two or more risk factors (odds ratio [OR] 2.91, 95% confidence interval [CI]: 1.09, 7.42) and pathologic T stage ≥T3 (OR 4.21, 95% CI: 1.13-15.8) were independently associated with a higher 90-d complication rate. The 3-yr estimated overall survival and recurrence-free survival rates were 82% and 47%, respectively. Limitations include the retrospective nature and heterogeneous cohort in terms of clinicopathologic characteristics and ICI regimens received. CONCLUSIONS Nephrectomy following ICI therapy is feasible and a potential consolidative therapy option in select patients. Further research in the neoadjuvant setting is also warranted. PATIENT SUMMARY This study evaluates the outcomes of kidney surgery following immune checkpoint inhibitor therapy (mainly nivolumab and ipilimumab or pembrolizumab and axitinib) for patients with advanced kidney cancer. We utilized data from five academic centers across the USA and found that surgery in this setting did not have more complications or returns to the hospital than similar surgeries, indicating that it is a safe and feasible procedure at this time.
Collapse
Affiliation(s)
- Wesley Yip
- University of Southern California, Los Angeles, CA, USA
| | | | | | - Randall Lee
- Fox Chase Cancer Center - Temple Health, Philadelphia, PA, USA
| | | | - Aeen Asghar
- University of Southern California, Los Angeles, CA, USA; Fox Chase Cancer Center - Temple Health, Philadelphia, PA, USA
| | | | - Jie Cai
- University of Southern California, Los Angeles, CA, USA
| | - Manju Aron
- University of Southern California, Los Angeles, CA, USA
| | - Inderbir Gill
- University of Southern California, Los Angeles, CA, USA
| | | | - Robert Uzzo
- Fox Chase Cancer Center - Temple Health, Philadelphia, PA, USA
| | | | | | | |
Collapse
|
3
|
Thi HV, Ngo AD, Tran LT, Chu DT. Phage for cancer therapy. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 201:225-239. [PMID: 37770174 DOI: 10.1016/bs.pmbts.2023.03.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
Cancer is currently a global health challenge, characterized by dysfunction of organs due to the uncontrolled growth of cells exponentially. The therapies used to treat cancer in patients so far are widely used. However, there are also some problems, such as the high cost of surgery and chemotherapy. Thus, there are many barriers to care for patients with cancer, especially in low and middle-income countries. In addition, the many risks and adverse effects of radiation treatment. Therefore, to reduce mortality in patients with the disease, we need a newer therapy with more targeted treatment, fewer side effects, and cheaper cost. The application of bacteria in cancer treatment was first developed in 1983. Currently, this therapy is attracting the attention of scientists due to its great potential in cancer treatment. This chapter discusses the successful research on the bacteriophage for cancer, the mechanism and its potential. In addition, some types of bacteria that are most important for cancer treatment and limitations on the widespread application of this therapy were also mentioned. Reviewing all the researches on bacteriotherapy in cancer are essential to increase the knowledge in this area and make this therapy more optimal in the future.
Collapse
Affiliation(s)
- Hue Vu Thi
- Center for Biomedicine and Community Health, International School, Vietnam National University, Hanoi, Vietnam; Faculty of Applied Sciences, International School, Vietnam National University, Hanoi, Vietnam
| | - Anh-Dao Ngo
- Center for Biomedicine and Community Health, International School, Vietnam National University, Hanoi, Vietnam
| | - Linh-Thao Tran
- Center for Biomedicine and Community Health, International School, Vietnam National University, Hanoi, Vietnam
| | - Dinh-Toi Chu
- Center for Biomedicine and Community Health, International School, Vietnam National University, Hanoi, Vietnam; Faculty of Applied Sciences, International School, Vietnam National University, Hanoi, Vietnam.
| |
Collapse
|
4
|
Wang Z, Zhu L, Li K, Sun Y, Giamas G, Stebbing J, Peng L, Yu Z. Alternative splicing events in tumor immune infiltration in renal clear cell carcinomas. Cancer Gene Ther 2022; 29:1418-1428. [PMID: 35370291 DOI: 10.1038/s41417-022-00426-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 11/23/2021] [Accepted: 01/07/2022] [Indexed: 11/09/2022]
Abstract
Alternative splicing (AS) is a gene regulatory mechanism that drives protein diversity and dysregulation of AS plays a significant role in tumorigenesis. This study aimed to develop a prognostic signature based on AS and elucidate the role in tumor immune microenvironment (TIME) in clear cell renal cell carcinoma (ccRCC). The prognosis-related AS events were analyzed by univariate Cox regression analysis. Gene set enrichment analyses (GSEA) were performed for functional annotation. Prognostic signatures were identified and validated using univariate and multivariate Cox regression, LASSO regression, Kaplan-Meier survival analysis, and proportional hazards model. The context of TIME in ccRCC was also analyzed. Gene and protein expression data of C4orf19 were obtained from ONCOMINE website and Human Protein Altas. Splicing factors (SFs) regulatory networks were visualized. 4431 survival-related AS events in ccRCC were screened. Based on splicing subtypes, eight AS prognostic signatures were constructed. A nomogram with good prognostic prediction was generated. Furthermore, the prognostic signatures were significantly correlated with TIME diversity and immune checkpoint inhibitor (ICI)-related genes. C4orf19 was the only gene whose expression levels were downregulated among the prognostic AS-related genes, which is considered as a promising prognostic factor in ccRCC. Potential functions of SFs were determined by splicing regulatory networks. In our study, AS patterns of novel indicators for prognostic prediction of ccRCC were explored. The AS-SF networks provide information of regulatory mechanisms. Players of AS events related to TIME were investigated, which contribute to prognosis monitoring of ccRCC.
Collapse
Affiliation(s)
- Zhiqiang Wang
- Department of Urology, Shouguang Hospital of Traditional Chinese Medicine, Shouguang, Shandong Province, China
| | - Liping Zhu
- Department of Medical Oncology, Shouguang Hospital of Traditional Chinese Medicine, Shouguang, Shandong Province, China
| | - Kesang Li
- Department of Hematology and Oncology, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, 315000, Zhejiang Province, China
| | - Yilan Sun
- Department of Respiratory Disease, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang Province, China
| | - Georgios Giamas
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, Brighton, UK
| | - Justin Stebbing
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, London, UK
| | - Ling Peng
- Department of Respiratory Disease, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang Province, China.
| | - Zhentao Yu
- Department of Thoracic Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital and Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China.
| |
Collapse
|
5
|
Wang S, Zhao Z, Xie J, Ren S, Tang S, Lin F, Qu J, Liu L. Accurate evaluation of the treatment effects of immunotherapy on subcutaneous ovarian cancer in mice with nonlinear optical imaging and algorithmic analysis. BIOMEDICAL OPTICS EXPRESS 2022; 13:2266-2277. [PMID: 35519272 PMCID: PMC9045922 DOI: 10.1364/boe.452569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 02/05/2022] [Accepted: 03/10/2022] [Indexed: 06/14/2023]
Abstract
Immunotherapy and its evaluation have shown great promise for cancer treatment. Here, a mouse subcutaneous transplantable tumor model was applied to testing therapeutic strategies. The mouse model was treated by regulating anti-PD-L1, anti-CTLA-4, cisplatin and their combined therapy. Biochemistry experiments have found that after immunotherapy, mice have more immune responses, which were manifested by an increase in the content of growth factors and the activation of T cells. Meanwhile, multimodal nonlinear optical microscopy imaging combined with algorithms was used to evaluate the treatment's effectiveness. By detecting the metabolism rate and microstructure in tissue, it was proved that combined therapies including immune checkpoint inhibitors do have a better effect on ovarian tumors. Our discovery of valid treatments for mice with ovarian tumor and provides an evaluation tool via nonlinear optics combined with algorithms offers new insights into therapeutic effect.
Collapse
Affiliation(s)
- Shiqi Wang
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic Engineering, Shenzhen University, China
- These authors contributed equally
| | - Zewei Zhao
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic Engineering, Shenzhen University, China
- These authors contributed equally
| | - Jiajun Xie
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic Engineering, Shenzhen University, China
| | - Sheng Ren
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic Engineering, Shenzhen University, China
| | - Shiyao Tang
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic Engineering, Shenzhen University, China
| | - Fangrui Lin
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic Engineering, Shenzhen University, China
| | - Junle Qu
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic Engineering, Shenzhen University, China
| | - Liwei Liu
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic Engineering, Shenzhen University, China
| |
Collapse
|
6
|
Zhang P, Meng J, Li Y, Yang C, Hou Y, Tang W, McHugh KJ, Jing L. Nanotechnology-enhanced immunotherapy for metastatic cancer. Innovation (N Y) 2021; 2:100174. [PMID: 34766099 PMCID: PMC8571799 DOI: 10.1016/j.xinn.2021.100174] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 10/11/2021] [Indexed: 12/14/2022] Open
Abstract
A vast majority of cancer deaths occur as a result of metastasis. Unfortunately, effective treatments for metastases are currently lacking due to the difficulty of selectively targeting these small, delocalized tumors distributed across a variety of organs. However, nanotechnology holds tremendous promise for improving immunotherapeutic outcomes in patients with metastatic cancer. In contrast to conventional cancer immunotherapies, rationally designed nanomaterials can trigger specific tumoricidal effects, thereby improving immune cell access to major sites of metastasis such as bone, lungs, and lymph nodes, optimizing antigen presentation, and inducing a persistent immune response. This paper reviews the cutting-edge trends in nano-immunoengineering for metastatic cancers with an emphasis on different nano-immunotherapeutic strategies. Specifically, it discusses directly reversing the immunological status of the primary tumor, harnessing the potential of peripheral immune cells, preventing the formation of a pre-metastatic niche, and inhibiting the tumor recurrence through postoperative immunotherapy. Finally, we describe the challenges facing the integration of nanoscale immunomodulators and provide a forward-looking perspective on the innovative nanotechnology-based tools that may ultimately prove effective at eradicating metastatic diseases.
Collapse
Affiliation(s)
- Peisen Zhang
- Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Bei Yi Jie 2, Zhong Guan Cun, Beijing 100190, China
| | - Junli Meng
- Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Bei Yi Jie 2, Zhong Guan Cun, Beijing 100190, China
| | - Yingying Li
- Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Bei Yi Jie 2, Zhong Guan Cun, Beijing 100190, China
| | - Chen Yang
- Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Bei Yi Jie 2, Zhong Guan Cun, Beijing 100190, China
| | - Yi Hou
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Wen Tang
- South China Advanced Institute for Soft Matter Science and Technology, State Key Laboratory of Luminescent Materials and Devices, South China University of Technology, Guangzhou 510640, China
| | - Kevin J McHugh
- Department of Bioengineering, Rice University, 6100 Main Street, MS-142, Houston, TX 77005, USA
| | - Lihong Jing
- Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Bei Yi Jie 2, Zhong Guan Cun, Beijing 100190, China
| |
Collapse
|
7
|
Qiu Y, Wang X, Fan Z, Zhan S, Jiang X, Huang J. Integrated analysis on the N6-methyladenosine-related long noncoding RNAs prognostic signature, immune checkpoints, and immune cell infiltration in clear cell renal cell carcinoma. IMMUNITY INFLAMMATION AND DISEASE 2021; 9:1596-1612. [PMID: 34432955 PMCID: PMC8589390 DOI: 10.1002/iid3.513] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 07/27/2021] [Accepted: 08/13/2021] [Indexed: 12/17/2022]
Abstract
Background Patients with advanced clear cell renal cell carcinoma (ccRCC) have a poor prognosis and lack effective prognostic biomarkers. N6‐methyladenosine‐related lncRNAs (m6A‐related long noncoding RNAs [lncRNAs]) have been confirmed to be associated with the development of multiple tumors, but its role in ccRCC is not clear. Methods Gene expression data and clinical information of ccRCC patients were extracted from The Cancer Genome Atlas Database. The prognostic m6A‐related lncRNAs were obtained by Pearson's correlation analysis and univariate Cox regression analysis. Afterward, the cluster classification and its correlation with prognosis, clinical characteristics, and immunity were analyzed. LASSO regression was used to establish the prognostic risk model. The predictive performance of the prognostic model was evaluated and validated by survival analysis and receiver operating characteristic curve analysis, et al. The expression of immune checkpoints and immune cell infiltration in patients with different risks were systematically analyzed. Results A total of 27 prognostic m6A‐related lncRNAs were identified. These m6A‐related lncRNAs were differentially expressed between tumor and normal tissues. Among them, 24 high‐risk m6A‐related lncRNAs were overexpressed in Cluster 2 and correlated with poor prognosis, low stromal score, high expression of immune checkpoints, and immunosuppressive cells infiltration. Based upon, a prognostic risk model composed of seven m6A‐related lncRNAs was constructed. After a series of analyses, it was proved that this model had good sensitivity and specificity, and could predict the prognosis of patients with different clinical stratification. The expression of PD‐1, PD‐L1, CTLA‐4, LAG‐3, TIM‐3, and TIGIT were significantly increased in the high‐risk patients, and there was a correlation between the risk score and immune cell infiltration. Conclusions The seven m6A‐related lncRNAs prognostic risk signature showed reliable prognostic predictive power for ccRCC and was associated with the expression of immune checkpoints and immune cell infiltration. This seven m6A‐related lncRNAs signature will be helpful in managing ccRCC and guiding individualized immunotherapy.
Collapse
Affiliation(s)
- Yuqin Qiu
- Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaogang Wang
- Department of Emergency Medicine, Beijing Haidian Hospital, Haidian Section of Peking University Third Hospital, Beijing, China
| | - Zhenjia Fan
- Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Shanhui Zhan
- Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xin Jiang
- Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jinchang Huang
- Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China.,Institute of Acupuncture and Moxibustion in Cancer Care, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
8
|
Ottaviano M, Curvietto M, Rescigno P, Tortora M, Palmieri G, Giannarelli D, Aieta M, Assalone P, Attademo L, Avallone A, Bloise F, Bosso D, Borzillo V, Buono G, Calderoni G, Caputo F, Cartenì G, Cavallero D, Cavo A, Ciardiello F, Conca R, Conteduca V, De Falco S, De Felice M, De Laurentiis M, De Placido P, De Placido S, De Santo I, De Stefano A, Della Corte CM, Di Franco R, Di Lauro V, Fabbrocini A, Federico P, Festino L, Giordano P, Giuliano M, Gridelli C, Grimaldi AM, Lia M, Marretta AL, Massa V, Mennitto A, Merler S, Merz V, Messina C, Messina M, Milano M, Minisini AM, Montesarchio V, Morabito A, Morgillo F, Mucci B, Nappi L, Napolitano F, Paciolla I, Pagliuca M, Palmieri G, Parola S, Pepe S, Petrillo A, Piantedosi F, Piccin L, Picozzi F, Pietroluongo E, Pignata S, Prati V, Riccio V, Rosanova M, Rossi A, Russo A, Salati M, Santabarbara G, Sbrana A, Simeone E, Silvestri A, Spada M, Tarantino P, Taveggia P, Tomei F, Vincenzo T, Trapani D, Trojanello C, Vanella V, Vari S, Ventriglia J, Vitale MG, Vitiello F, Vivaldi C, von Arx C, Zacchi F, Zampiva I, Zivi A, Daniele B, Ascierto PA. Impact of COVID-19 outbreak on cancer immunotherapy in Italy: a survey of young oncologists. J Immunother Cancer 2020; 8:jitc-2020-001154. [PMID: 33060148 PMCID: PMC7565202 DOI: 10.1136/jitc-2020-001154] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/08/2020] [Indexed: 01/20/2023] Open
Abstract
Background The coronavirus disease 2019 (COVID-19) pandemic has overwhelmed the health systems worldwide. Data regarding the impact of COVID-19 on cancer patients (CPs) undergoing or candidate for immune checkpoint inhibitors (ICIs) are lacking. We depicted the practice and adaptations in the management of patients with solid tumors eligible or receiving ICIs during the COVID-19 pandemic, with a special focus on Campania region. Methods This survey (25 questions), promoted by the young section of SCITO (Società Campana di ImmunoTerapia Oncologica) Group, was circulated among Italian young oncologists practicing in regions variously affected by the pandemic: high (group 1), medium (group 2) and low (group 3) prevalence of SARS-CoV-2–positive patients. For Campania region, the physician responders were split into those working in cancer centers (CC), university hospitals (UH) and general hospitals (GH). Percentages of agreement, among High (H) versus Medium (M) and versus Low (L) group for Italy and among CC, UH and GH for Campania region, were compared by using Fisher’s exact tests for dichotomous answers and χ2 test for trends relative to the questions with 3 or more options. Results This is the first Italian study to investigate the COVID-19 impact on cancer immunotherapy, unique in its type and very clear in the results. The COVID-19 pandemic seemed not to affect the standard practice in the prescription and delivery of ICIs in Italy. Telemedicine was widely used. There was high consensus to interrupt immunotherapy in SARS-CoV-2–positive patients and to adopt ICIs with longer schedule interval. The majority of the responders tended not to delay the start of ICIs; there were no changes in supportive treatments, but some of the physicians opted for delaying surgeries (if part of patients’ planned treatment approach). The results from responders in Campania did not differ significantly from the national ones. Conclusion Our study highlights the efforts of Italian oncologists to maintain high standards of care for CPs treated with ICIs, regardless the regional prevalence of COVID-19, suggesting the adoption of similar solutions. Research on patients treated with ICIs and experiencing COVID-19 will clarify the safety profile to continue the treatments, thus informing on the most appropriate clinical conducts.
Collapse
Affiliation(s)
- Margaret Ottaviano
- Oncology Unit, Ospedale del Mare, Napoli, Campania, Italy.,Department of Clinical Medicine and Surgery, Università degli Studi di Napoli Federico II and CRCTR Coordinating Rare Tumors Reference Center of Campania Region, AOU Federico II, Napoli, Italy
| | - Marcello Curvietto
- Unit of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Campania, Italy
| | - Pasquale Rescigno
- Clinical Studies, The Institute of Cancer Research and the Royal Marsden NHS Foundation Trust, London, UK
| | - Marianna Tortora
- CRCTR Coordinating Rare Tumors Reference Center of Campania Region, AOU Federico II, Napoli, Campania, Italy
| | - Giovannella Palmieri
- CRCTR Coordinating Rare Tumors Reference Center of Campania Region, AOU Federico II, Napoli, Campania, Italy
| | - Diana Giannarelli
- Biostatistics Unit, IRCCS Regina Elena National Cancer Institute, Roma, Lazio, Italy
| | - Michele Aieta
- Medical Oncology Unit, Department of Onco-Hematology, IRCCS-CROB Referral Cancer Center of Basilicata, Rionero in Vulture, Basilicata, Italy
| | - Pasquale Assalone
- Oncology Unit, Ospedale Ferdinando Veneziale, Isernia, Molise, Italy
| | - Laura Attademo
- Department of Urology and Gynecology, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Campania, Italy
| | - Antonio Avallone
- Abdominal Oncology Unit, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Campania, Italy
| | | | | | - Valentina Borzillo
- Department of Radioterapia, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Campania, Italy
| | - Giuseppe Buono
- Oncology Unit, San Rocco Hospital, Sessa Aurunca, Campania, Italy
| | | | - Francesca Caputo
- U.O.C. Oncologia, Azienda Ospedaliera dei Colli, Monaldi Hospital, Napoli, Campania, Italy
| | - Giacomo Cartenì
- Department of Medical Oncology, AORN "A. Cardarelli", Napoli, Campania, Italy
| | | | - Alessia Cavo
- Oncology Unit, Villa Scassi Hospital, Genova, Italy
| | - Fortunato Ciardiello
- Oncology, Department of Precision Medicine, Università della Campania "L. Vanvitelli", Napoli, Campania, Italy
| | - Raffaele Conca
- Medical Oncology Unit, Department of Onco-Hematology, IRCCS-CROB Referral Cancer Center of Basilicata, Rionero in Vulture, Basilicata, Italy
| | - Vincenza Conteduca
- Medical Oncology Department, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Emilia-Romagna, Italy
| | - Stefano De Falco
- Oncology Unit, Sant'Ottone Frangipane Hospital, Avellino, Campania, Italy
| | - Marco De Felice
- Oncology, Department of Precision Medicine, Università della Campania "L. Vanvitelli", Napoli, Campania, Italy.,Oncology Unit, Sant'Anna e San Sebastiano, Caserta, Campania, Italy
| | | | - Pietro De Placido
- Department of Clinical Medicine and Surgery, Oncology Unit, Università degli Studi di Napoli Federico II, Napoli, Campania, Italy
| | - Sabino De Placido
- CRCTR Coordinating Rare Tumors Reference Center of Campania Region, AOU Federico II, Napoli, Campania, Italy.,Department of Clinical Medicine and Surgery, Oncology Unit, Università degli Studi di Napoli Federico II, Napoli, Campania, Italy
| | - Irene De Santo
- Oncology Unit, Gemelli Molise s.p.a, Campobasso, Molise, Italy
| | - Alfonso De Stefano
- Abdominal Oncology Unit, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Campania, Italy
| | - Carminia Maria Della Corte
- Oncology, Department of Precision Medicine, Università della Campania "L. Vanvitelli", Napoli, Campania, Italy
| | - Rossella Di Franco
- Department of Radioterapia, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Campania, Italy
| | - Vincenzo Di Lauro
- Breast Unit, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Campania, Italy.,Department of Clinical Medicine and Surgery, Oncology Unit, Università degli Studi di Napoli Federico II, Napoli, Campania, Italy
| | | | | | - Lucia Festino
- Unit of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Campania, Italy
| | | | - Mario Giuliano
- CRCTR Coordinating Rare Tumors Reference Center of Campania Region, AOU Federico II, Napoli, Campania, Italy.,Department of Clinical Medicine and Surgery, Oncology Unit, Università degli Studi di Napoli Federico II, Napoli, Campania, Italy
| | - Cesare Gridelli
- Division of Medical Oncology, "San Giuseppe Moscati" Hospital, Avellino, Campania, Italy
| | - Antonio Maria Grimaldi
- Unit of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Campania, Italy
| | - Michela Lia
- Oncology Unit, SS Antonio e Biagio e Cesare Arrigo Hospital, Alessandria, Piemonte, Italy
| | - Antonella Lucia Marretta
- Department of Clinical Medicine and Surgery, Oncology Unit, Università degli Studi di Napoli Federico II, Napoli, Campania, Italy
| | - Valentina Massa
- Medical Oncology Unit, University of Pisa, Pisa, Toscana, Italy
| | - Alessia Mennitto
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Lombardia, Italy
| | - Sara Merler
- Department of Oncology, University of Verona, Verona, Veneto, Italy
| | - Valeria Merz
- Department of Medical Oncology, Santa Chiara Hospital, Trento, Trentino Alto Adige, Italy
| | - Carlo Messina
- Department of Medical Oncology, Santa Chiara Hospital, Trento, Trentino Alto Adige, Italy
| | - Marco Messina
- UOC Oncologia Medica, Ospedali Riuniti Villa Sofia Cervello, Palermo, Sicilia, Italy
| | - Monica Milano
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Lombardia, Italy
| | - Alessandro Marco Minisini
- Department of Oncology, Azienda Sanitaria Universitaria Integrata del Friuli Centrale, Udine, Friuli Venezia Giulia, Italy
| | - Vincenzo Montesarchio
- U.O.C. Oncologia, Azienda Ospedaliera dei Colli, Monaldi Hospital, Napoli, Campania, Italy
| | - Alessandro Morabito
- Thoracic Medical Oncology, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Campania, Italy
| | - Floriana Morgillo
- Oncology, Department of Precision Medicine, Università della Campania "L. Vanvitelli", Napoli, Campania, Italy
| | - Brigitta Mucci
- Department of Clinical Medicine and Surgery, Oncology Unit, Università degli Studi di Napoli Federico II, Napoli, Campania, Italy
| | - Lucia Nappi
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada and British Columbia Cancer Agency-Vancouver Center-Department of Medicine, Division of Medical Oncology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Fabiana Napolitano
- Department of Clinical Medicine and Surgery, Oncology Unit, Università degli Studi di Napoli Federico II, Napoli, Campania, Italy
| | - Immacolata Paciolla
- Oncology Unit, Ospedale Sacro Cuore di Gesù Fatebenefratelli, Benevento, Campania, Italy
| | - Martina Pagliuca
- Department of Clinical Medicine and Surgery, Oncology Unit, Università degli Studi di Napoli Federico II, Napoli, Campania, Italy
| | - Giuseppe Palmieri
- Institute of Research on Genetics and Biomedicine (IRGB), National Research Council (CNR), Sassari, Sardegna, Italy
| | - Sara Parola
- Department of Clinical Medicine and Surgery, Oncology Unit, Università degli Studi di Napoli Federico II, Napoli, Campania, Italy
| | - Stefano Pepe
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", Oncology Unit, University of Salerno, Baronissi, Campania, Italy
| | - Angelica Petrillo
- Oncology Unit, Ospedale del Mare, Napoli, Italy.,Oncology, Department of Precision Medicine, Università della Campania "L. Vanvitelli", Napoli, Campania, Italy
| | - Francovito Piantedosi
- U.O.C. Pneumologia Oncologica, Azienda Ospedaliera dei Colli, Monaldi Hospital, Naples, Campania, Italy
| | - Luisa Piccin
- Veneto Institute of Oncology IOV-IRCCS, Padova, Veneto, Italy
| | - Fernanda Picozzi
- Department of Clinical Medicine and Surgery, Oncology Unit, Università degli Studi di Napoli Federico II, Napoli, Campania, Italy
| | - Erica Pietroluongo
- Department of Clinical Medicine and Surgery, Oncology Unit, Università degli Studi di Napoli Federico II, Napoli, Campania, Italy
| | - Sandro Pignata
- Department of Urology and Gynecology, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Campania, Italy
| | | | - Vittorio Riccio
- Department of Clinical Medicine and Surgery, Oncology Unit, Università degli Studi di Napoli Federico II, Napoli, Campania, Italy
| | | | - Alice Rossi
- Department of Oncology, University of Verona, Verona, Veneto, Italy
| | - Anna Russo
- Medical Oncology Unit, Fondazione IRCCS Giovanni Paolo II, Bari, Puglia, Italy
| | - Massimiliano Salati
- Medical Oncology Unit, University Hospital Modena, Modena, Emilia Romagna, Italy
| | - Giuseppe Santabarbara
- Division of Medical Oncology, "San Giuseppe Moscati" Hospital, Avellino, Campania, Italy
| | - Andrea Sbrana
- Medical Oncology Unit, University of Pisa, Pisa, Toscana, Italy
| | - Ester Simeone
- Unit of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Campania, Italy
| | | | - Massimiliano Spada
- Oncology Unit, Fondazione Istituto San Raffaele G Giglio di Cefalu, Cefalu, Sicilia, Italy
| | - Paolo Tarantino
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milano, Lombardia, Italy
| | | | - Federica Tomei
- Oncology Unit, Ospedale Ferdinando Veneziale, Isernia, Molise, Italy
| | - Tortora Vincenzo
- Department of Clinical Medicine and Surgery, Oncology Unit, Università degli Studi di Napoli Federico II, Napoli, Campania, Italy.,Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", Oncology Unit, University of Salerno, Baronissi, Campania, Italy
| | - Dario Trapani
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milano, Lombardia, Italy
| | - Claudia Trojanello
- Unit of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Campania, Italy
| | - Vito Vanella
- Unit of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Campania, Italy
| | - Sabrina Vari
- Medical Oncology 1 Unit, IRCCS Regina Elena National Cancer Institute, Rome, Lazio, Italy
| | - Jole Ventriglia
- Oncology Division, ASL San Felice a Cancello, Caserta, Campania, Italy
| | - Maria Grazia Vitale
- Unit of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Campania, Italy.,Department of Oncology, Azienda Sanitaria Universitaria Integrata del Friuli Centrale, Udine, Friuli Venezia Giulia, Italy
| | - Fabiana Vitiello
- U.O.C. Pneumologia Oncologica, Azienda Ospedaliera dei Colli, Monaldi Hospital, Naples, Campania, Italy
| | | | - Claudia von Arx
- Department of Surgery and Cancer, Imperial College, London, UK
| | - Francesca Zacchi
- Oncology Unit, Università degli Studi di Verona, sede Borgo Roma, Verona, Veneto, Italy
| | - Ilaria Zampiva
- Oncology Unit, Università degli Studi di Verona, sede Borgo Roma, Verona, Veneto, Italy
| | - Andrea Zivi
- Department of Surgery and Cancer, Imperial College, London, UK.,Medical Oncology Department, AOUI di Verona, Verona, Veneto, Italy
| | | | - Paolo Antonio Ascierto
- Unit of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Campania, Italy
| | | |
Collapse
|
9
|
Singla N, Hutchinson RC, Ghandour RA, Freifeld Y, Fang D, Sagalowsky AI, Lotan Y, Bagrodia A, Margulis V, Hammers HJ, Woldu SL. Improved survival after cytoreductive nephrectomy for metastatic renal cell carcinoma in the contemporary immunotherapy era: An analysis of the National Cancer Database. Urol Oncol 2020; 38:604.e9-604.e17. [PMID: 32253116 PMCID: PMC7269798 DOI: 10.1016/j.urolonc.2020.02.029] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 02/23/2020] [Accepted: 02/29/2020] [Indexed: 12/22/2022]
Abstract
OBJECTIVES Despite immune checkpoint inhibitor (ICI) approval for metastatic renal cell carcinoma (mRCC) in 2015, cytoreductive nephrectomy (CN) is guided by extrapolation from earlier classes of therapy. We evaluated survival outcomes, timing, and safety of combining CN with modern immunotherapy (IO) for mRCC. METHODS From 96,329 renal cancer cases reported to the NCDB between 2015 and 2016, we analyzed 391 surgical candidates diagnosed with clear cell mRCC treated with IO ± CN and no other systemic therapies. Primary outcome was overall survival (OS) stratified by the performance of CN (CN + IO vs. IO alone). Secondary outcomes included OS stratified by the timing of CN, pathologic findings, and perioperative outcomes. RESULTS Of 391 patients, 221 (56.5%) received CN + IO and 170 (43.5%) received IO only. Across a median follow-up of 14.7 months, patients who underwent CN + IO had superior OS (median NR vs. 11.6 months; hazard ratio 0.23, P < 0.001), which was upheld on multivariable analyses. IO before CN resulted in lower pT stage, grade, tumor size, and lymphovascular invasion rates compared to upfront CN. Two of 20 patients (10%) undergoing CN post-IO achieved complete pathologic response in the primary tumor (pT0). There were no positive surgical margins, 30-day readmissions, or prolonged length of stay in patients undergoing delayed CN. CONCLUSION Using a large, national, registry-based cohort, we provide the first report of survival outcomes in mRCC patients treated with CN combined with modern IO. Our findings support an oncologic role for CN in the ICI era and provide preliminary evidence regarding the timing and safety of CN relative to IO administration.
Collapse
Affiliation(s)
- Nirmish Singla
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX; Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ryan C Hutchinson
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Rashed A Ghandour
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Yuval Freifeld
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Dong Fang
- Department of Urology, Peking University First Hospital, Institute of Urology, Peking University, National Urological Cancer Center, Beijing, China
| | - Arthur I Sagalowsky
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Yair Lotan
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Aditya Bagrodia
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Vitaly Margulis
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX; Department of Urology, I.M. Sechenov First Moscow State University, Moscow, Russia
| | - Hans J Hammers
- Division of Hematology and Oncology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX
| | - Solomon L Woldu
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX.
| |
Collapse
|
10
|
Singla N, Ghandour RA, Margulis V. Is cytoreductive nephrectomy relevant in the immunotherapy era? Curr Opin Urol 2020; 29:526-530. [PMID: 31305273 DOI: 10.1097/mou.0000000000000659] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW The recent approval of immune checkpoint inhibitors (ICI) revolutionized the treatment paradigm for metastatic renal cell carcinoma (mRCC). However, the role of cytoreductive nephrectomy is not well defined in this setting. Here, we review the contemporary role and timing of cytoreductive nephrectomy for advanced RCC in the era of immunotherapy. RECENT FINDINGS Evidence concerning combination systemic therapy and cytoreductive nephrectomy in the multimodal management of mRCC is primarily limited to studies conducted in the targeted therapy era. The oncologic role of cytoreductive nephrectomy in the setting of ICI remains largely undefined and is supported primarily by case reports. Nonetheless, patient selection for cytoreductive nephrectomy is paramount, and appropriate candidacy in the ICI era will likely be refined as increasing evidence emerges. Until then, a cautious balance between perceived oncologic benefit and risks of intervention must be exercised. Several trials are ongoing to help shed light on patient selection, technical feasibility, and optimal timing of cytoreductive nephrectomy in the immunotherapy era. SUMMARY Although the role and timing for cytoreductive nephrectomy remain to be further elucidated in the immunotherapy era, patient selection remains critical for treatment planning. Further studies are urgently needed to better define the role of cytoreductive nephrectomy in this setting.
Collapse
Affiliation(s)
- Nirmish Singla
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | | | | |
Collapse
|
11
|
Singla N, Elias R, Ghandour RA, Freifeld Y, Bowman IA, Rapoport L, Enikeev M, Lohrey J, Woldu SL, Gahan JC, Bagrodia A, Brugarolas J, Hammers HJ, Margulis V. Pathologic response and surgical outcomes in patients undergoing nephrectomy following receipt of immune checkpoint inhibitors for renal cell carcinoma. Urol Oncol 2019; 37:924-931. [DOI: 10.1016/j.urolonc.2019.08.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 06/26/2019] [Accepted: 08/19/2019] [Indexed: 01/16/2023]
|
12
|
Rational Approaches to Treatment Duration with Immunotherapy in Metastatic Renal Cell Carcinoma. Eur Urol Focus 2019; 6:31-33. [PMID: 31757714 DOI: 10.1016/j.euf.2019.10.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 10/17/2019] [Indexed: 01/28/2023]
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized the treatment paradigm for metastatic renal cell carcinoma. The appropriate duration for ICI treatment is not clear, however. Analyses of landmark trials reveal that some patients exhibit sustained durable responses to ICIs even after treatment discontinuation, resulting in prolonged treatment-free intervals that can mitigate potential toxicities and the considerable financial burden associated with treatment. Adaptive approaches with PD1 monotherapy and combination immunotherapy tailored to tumor response are ongoing. More efforts will be needed to clarify the ideal ICI dosing regimen to maximize oncological benefit while minimizing treatment-related adverse effects and costs. PATIENT SUMMARY: We reviewed considerations surrounding treatment strategies when using immunotherapy to treat patients with kidney cancer. It is clear that some patients can experience prolonged cancer control when discontinuing immunotherapy. However, individualized approaches will be necessary to strike a balance between optimizing patient outcomes and reducing unnecessary side effects and cost.
Collapse
|
13
|
Limitations to the Therapeutic Potential of Tyrosine Kinase Inhibitors and Alternative Therapies for Kidney Cancer. Ochsner J 2019; 19:138-151. [PMID: 31258426 DOI: 10.31486/toj.18.0015] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background: Renal cell carcinomas (RCCs) are the most common primary renal tumor. RCCs have a high rate of metastasis and have the highest mortality rate of all genitourinary cancers. They are often diagnosed late when metastases have developed, and these metastases are difficult to treat successfully. Since 2006, the standard first-line treatment for patients with metastatic RCC has been multitargeted tyrosine kinase inhibitors (TKIs) that include mammalian target of rapamycin (mTOR) inhibitors. RCCs are highly vascularized tumors, and their angiogenesis is controlled by tyrosine kinases that play a vital role in growth factor signaling to stimulate this process. TKI therapy was introduced for direct targeting of angiogenesis in RCC. TKIs have been moderately successful in the treatment of metastatic RCC and initially increased cancer-specific survival times. However, RCC rapidly becomes resistant to TKIs, and no current drug has produced a cure for advanced RCC. Methods: We provide an overview of RCC, explain some reasons for therapy resistance in RCC, and describe some therapies that may overcome resistance to TKIs. The key pathways that determine therapy resistance are illustrated. Results: Factors involved in the development and progression of RCC include genetic mutations, activation of hypoxia-inducible factor and related proteins, cellular metabolism, the tumor microenvironment, and growth factors and their receptors. Resistance to the therapeutic potential of TKIs can be acquired or intrinsic. Alternative therapies include other small molecule drugs and immunotherapy based on immune checkpoint blockade. Conclusion: The treatment of RCC is undergoing a paradigm shift from sole use of small molecule antiangiogenesis TKIs as first-line therapy to include newly approved agents for second-line and third-line therapy that now involve the mTOR pathway and immune checkpoint blockade drugs for patients with advanced RCC.
Collapse
|
14
|
Ghandour RA, Singla N, Margulis V. The use of cytoreductive nephrectomy in patients with renal cell carcinoma. Expert Rev Anticancer Ther 2019; 19:405-411. [PMID: 31020871 DOI: 10.1080/14737140.2019.1606716] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION The systemic options for managing metastatic renal cell carcinoma (mRCC) have expanded considerably over the past decade. Initially limited to cytokines, clinicians may now choose from several classes of targeted therapies and, most recently, immune checkpoint inhibitors. Areas covered: In this review, we discuss the role and timing of cytoreductive nephrectomy (CN) and its evolution starting with cytokines, and then alongside the emergence of targeted therapy and novel immunotherapy with immune checkpoint inhibitors. Patient selection remains the most critical determinant in offering CN, and the anticipated survival benefits of CN must be weighed against the surgical morbidity and potential delay to receipt of systemic therapies. Expert opinion: Proper patient selection is key for decision-making in mRCC. Prospective data is urgently needed to define the role of CN in the contemporary immunotherapy era, with greater personalization of prognostic models.
Collapse
Affiliation(s)
- Rashed A Ghandour
- a Department of Urology , University of Texas Southwestern Medical Center , Dallas , TX , USA
| | - Nirmish Singla
- a Department of Urology , University of Texas Southwestern Medical Center , Dallas , TX , USA
| | - Vitaly Margulis
- a Department of Urology , University of Texas Southwestern Medical Center , Dallas , TX , USA
| |
Collapse
|
15
|
Graham K, Unger E. Overcoming tumor hypoxia as a barrier to radiotherapy, chemotherapy and immunotherapy in cancer treatment. Int J Nanomedicine 2018; 13:6049-6058. [PMID: 30323592 PMCID: PMC6177375 DOI: 10.2147/ijn.s140462] [Citation(s) in RCA: 378] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Hypoxia exists to some degree in most solid tumors due to inadequate oxygen delivery of the abnormal vasculature which cannot meet the demands of the rapidly proliferating cancer cells. The levels of oxygenation within the same tumor are highly variable from one area to another and can change over time. Tumor hypoxia is an important impediment to effective cancer therapy. In radiotherapy, the primary mechanism is the creation of reactive oxygen species; hypoxic tumors are therefore radiation resistant. A number of chemotherapeutic drugs have been shown to be less effective when exposed to a hypoxic environment which can lead to further disease progression. Hypoxia is also a potent barrier to effective immunotherapy in cancer treatment. Because of the recognition of hypoxia as an important barrier to cancer treatment, a variety of approaches have been undertaken to overcome or reverse tumor hypoxia. Such approaches have included breathing hyperbaric oxygen, artificial hemoglobins, allosteric hemoglobin modifiers, hypoxia activated prodrugs and fluorocarbons (FCs). These approaches have largely failed due to limited efficacy and/or adverse side effects. Oxygen therapeutics, based on liquid FCs, can potentially increase the oxygen-carrying capacity of the blood to reverse tumor hypoxia. Currently, at least two drugs are in clinical trials to reverse tumor hypoxia; one of these is designed to improve permeability of oxygen into the tumor tissue and the other is based upon a low boiling point FC that transports higher amounts of oxygen per gram than previously tested FCs.
Collapse
|
16
|
Chivu-Economescu M, Matei L, Necula LG, Dragu DL, Bleotu C, Diaconu CC. New therapeutic options opened by the molecular classification of gastric cancer. World J Gastroenterol 2018; 24:1942-1961. [PMID: 29760539 PMCID: PMC5949709 DOI: 10.3748/wjg.v24.i18.1942] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 04/12/2018] [Accepted: 04/23/2018] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer (GC) is one of the most lethal and aggressive cancers, being the third cause of cancer related death worldwide. Even with radical gastrectomy and the latest generation of molecular chemotherapeutics, the numbers of recurrence and mortality remains high. This is due to its biological heterogeneity based on the interaction between multiple factors, from genomic to environmental factors, diet or infections with various pathogens. Therefore, understanding the molecular characteristics at a genomic level is critical to develop new treatment strategies. Recent advances in GC molecular classification provide the unique opportunity to improve GC therapy by exploiting the biomarkers and developing novel targeted therapy specific to each subtype. This article highlights the molecular characteristics of each subtype of gastric cancer that could be considered in shaping a therapeutic decision, and also presents the completed and ongoing clinical trials addressed to those targets. The implementation of the novel molecular classification system will allow a preliminary patient selection for clinical trials, a mandatory issue if it is desired to test the efficacy of a certain inhibitor to the given target. This will represent a substantial advance as well as a powerful tool for targeted therapy. Nevertheless, translating the scientific results into new personalized treatment opportunities is needed in order to improve clinical care, the survival and quality of life of patients with GC.
Collapse
Affiliation(s)
- Mihaela Chivu-Economescu
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, Bucharest 030304, Romania
| | - Lilia Matei
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, Bucharest 030304, Romania
| | - Laura G Necula
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, Bucharest 030304, Romania
- Nicolae Cajal Institute, Titu Maiorescu University, Bucharest 040441, Romania
| | - Denisa L Dragu
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, Bucharest 030304, Romania
| | - Coralia Bleotu
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, Bucharest 030304, Romania
| | - Carmen C Diaconu
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, Bucharest 030304, Romania
| |
Collapse
|
17
|
Check point inhibitors a new era in renal cell carcinoma treatment. Med Oncol 2018; 35:85. [PMID: 29728867 DOI: 10.1007/s12032-018-1147-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Accepted: 05/02/2018] [Indexed: 12/31/2022]
Abstract
In the United States, the estimated number of new cases of renal cell carcinoma (RCC) is approximately 65,000 case with about 15,000 deaths in the year of 2018 (Siegel et al. in CA Cancer J Clin 68(1):7, 2018). RCC as an immunogenic malignancy is supported by many theories and facts which include tumor richness of lymphocytes infiltrate, the occurrence of spontaneous tumor regression, and the proved effect of traditional immunotherapy (Finke et al. in J Immunother 11(1):1-11, 1992), all these factors support the potential therapeutic effect of the novel immunotherapeutic agents in RCC. Historically, complete tumor regression in metastatic RCC is achievable in a minority of patients through traditional immunotherapies such as high-dose interleukin-2 (IL-2) (Fyfe et al. in J Clin Oncol 13(3):688, 1995) and interferon-alfa (IFNa) (Negrier et al. in N Engl J Med 338(18):1272, 1998); however due to the significant rate of toxicities and low efficacy; accordingly the targeted therapy with tyrosine kinase inhibitors (TKIs) and vascular endothelial growth factor-antibodies (VEGF) became the standard and prevalent treatment approach for advanced RCC both in front and subsequent lines of therapy (Escudier et al. in Ann Oncol. 25(Suppl 3):iii49-iii56, 2014). A new avenue of immunotherapy utilizing novel strategy to block immune checkpoints has emerged in a new era for RCC treatment (Ascierto et al. in J Transl Med 12:291, 2014). Results from clinical trials are encouraging in both front-line and second-line settings, in a phase III trial (CheckMate 025) nivolumab compared to everolimus improved overall survival in previously treated metastatic RCC who had progressed on prior treatment with targeting agents (Motzer et al. in N Engl J Med 373:1803, 2015). CheckMate 214, a phase III trial, demonstrated superior overall survival and objective response with combined checkpoint inhibitors compared to sunitinib in Treatment-Naïve Advanced RCC among intermediate- and poor-risk group (Motzer et al. in N Engl J Med. 378(14):1277-1290, 2018). In this review, we discuss the systemic Immunotherapy with checkpoint inhibitors that have been approved or are currently being investigated in RCC, clinical experience with these agents, and its future development.
Collapse
|
18
|
Antonelli A, Ferrari SM, Fallahi P. Current and future immunotherapies for thyroid cancer. Expert Rev Anticancer Ther 2017; 18:149-159. [PMID: 29241377 DOI: 10.1080/14737140.2018.1417845] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Cancer immunotherapies were approved in recent years, including immune checkpoint inhibitors. Experience with ipilimumab (CTLA-4 antagonist), nivolumab and pembrolizumab (PD-1 antagonists), and atezolizumab (PD-L1 antagonist) has shown that the impact on overall survival in cancer patients is paramount. Immune checkpoint inhibitors target the immune system and they can be applied across multiple cancers; the response rate is ranging from 20 to 40%. Many studies have shown that thyroid cancer (TC) cells produce cytokines and chemokines, inducing several tumor-promoting effects. Targeting and/or lowering cytokines and chemokines concentrations within the tumor microenvironment would produce a therapeutic benefit. In TC, increased Treg and PD-1+ T cell frequencies are indicative of aggressive disease and PD-L1 expression correlates with a greater risk of recurrence. Area covered: After performing a literature search, a few pioneering studies have evaluated immunotherapy in thyroid cancer. More recently a case has been described involving anaplastic thyroid cancer treated with vemurafenib and nivolumab, with substantial regression and complete radiographic and clinical remission. Expert commentary: The use of immune checkpoint inhibitors in aggressive TC has not yet been extensively investigated and further studies in a large number of TC patients are urgently needed.
Collapse
Affiliation(s)
- Alessandro Antonelli
- a Department of Clinical and Experimental Medicine , University of Pisa , Pisa , Italy
| | | | - Poupak Fallahi
- a Department of Clinical and Experimental Medicine , University of Pisa , Pisa , Italy
| |
Collapse
|
19
|
Basnet A, Khullar G, Mehta R, Chittoria N. A Case of Locally Advanced Castration-resistant Prostate Cancer With Remarkable Response to Nivolumab. Clin Genitourin Cancer 2017. [DOI: 10.1016/j.clgc.2017.05.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
20
|
Complete response to anti-PD-1 nivolumab in massive skin metastasis from melanoma. Anticancer Drugs 2017; 28:808-810. [DOI: 10.1097/cad.0000000000000515] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
21
|
Emens LA, Ascierto PA, Darcy PK, Demaria S, Eggermont AMM, Redmond WL, Seliger B, Marincola FM. Cancer immunotherapy: Opportunities and challenges in the rapidly evolving clinical landscape. Eur J Cancer 2017. [PMID: 28623775 DOI: 10.1016/j.ejca.2017.01.035] [Citation(s) in RCA: 390] [Impact Index Per Article: 48.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Cancer immunotherapy is now established as a powerful way to treat cancer. The recent clinical success of immune checkpoint blockade (antagonists of CTLA-4, PD-1 and PD-L1) highlights both the universal power of treating the immune system across tumour types and the unique features of cancer immunotherapy. Immune-related adverse events, atypical clinical response patterns, durable responses, and clear overall survival benefit distinguish cancer immunotherapy from cytotoxic cancer therapy. Combination immunotherapies that transform non-responders to responders are under rapid development. Current challenges facing the field include incorporating immunotherapy into adjuvant and neoadjuvant cancer therapy, refining dose, schedule and duration of treatment and developing novel surrogate endpoints that accurately capture overall survival benefit early in treatment. As the field rapidly evolves, we must prioritise the development of biomarkers to guide the use of immunotherapies in the most appropriate patients. Immunotherapy is already transforming cancer from a death sentence to a chronic disease for some patients. By making smart, evidence-based decisions in developing next generation immunotherapies, cancer should become an imminently treatable, curable and even preventable disease.
Collapse
Affiliation(s)
- Leisha A Emens
- Johns Hopkins University School of Medicine, Department of Oncology, Graduate Program in Pathobiology, Baltimore, MD 21287, USA.
| | - Paolo A Ascierto
- Istituto Nazionale Tumori Fondazione G. Pascale, Melanoma, Cancer Immunotherapy and Innovative Therapy Unit, Napoli, Italy
| | - Phillip K Darcy
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville 3010, Australia
| | - Sandra Demaria
- Weill Cornell Medical College, Department of Radiation Oncology, New York, NY 10065, USA
| | - Alexander M M Eggermont
- Cancer Institute Gustave-Roussy, 114 Rue Edouard Vaillant, Villejuif/Paris-Sud 94800, France
| | - William L Redmond
- Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute, Providence Portland Medical Center, Portland, OR 97213, USA
| | - Barbara Seliger
- Martin Luther University, Institute for Medical Immunology, Magdeburger Str. 2, 06112 Halle, Germany
| | | |
Collapse
|
22
|
Zhu X, Lang J. Programmed death-1 pathway blockade produces a synergistic antitumor effect: combined application in ovarian cancer. J Gynecol Oncol 2017; 28:e64. [PMID: 28657225 PMCID: PMC5540723 DOI: 10.3802/jgo.2017.28.e64] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 01/09/2017] [Accepted: 05/18/2017] [Indexed: 02/08/2023] Open
Abstract
Programmed death-1 (PD-1) and its ligand are part of the immune checkpoint pathway that down-regulates effector T cells in immune response, thereby causing immune suppression. The PD-1/programmed death-ligand 1 (PD-L1) pathway can be blocked by antibodies to reverse tumor-mediated immunosuppression. However, advanced cancers such as stage III-IV ovarian cancer (OC) and certain types such as ID8 OC (a clone of C57BL/6 mouse OC) may hijack the PD-1/PD-L1 pathway to escape immune attack. When combined with chemotherapy, radiotherapy, targeted therapy, immunotherapy, or other agents, these PD-1/PD-L1 pathway blockages can produce a synergistic antitumor response in OC. Combined immunotherapy significantly prolongs overall survival by changing the tumor microenvironment through processes such as increasing the number of CD4⁺ or CD8⁺ T cells or cytokines in mice with OC and decreasing the number of regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs). OC patients treated with combined immunotherapy received better prognoses than those treated with monotherapy. This review reflects the move toward novel therapy combinations for OC and discusses these promising immunotherapeutic approaches, which are more cost-effective and effective than other approaches.
Collapse
Affiliation(s)
- Xinxin Zhu
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Jinghe Lang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
23
|
Mazza C, Escudier B, Albiges L. Nivolumab in renal cell carcinoma: latest evidence and clinical potential. Ther Adv Med Oncol 2017; 9:171-181. [PMID: 28344662 PMCID: PMC5349425 DOI: 10.1177/1758834016679942] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Similar to melanoma, renal cell carcinoma (RCC) has been historically considered as an immunogenic tumor, with interleukin 2 (IL-2) and interferon alpha (IFN-α) being the first approved treatments in the 1990s. However, these therapies were effective in only 10-20% of cases and were not well tolerated. Recently, new insights on the interaction between the immune system and tumor have identified the programmed death-1/programmed death-ligand-1 (PD-1/PD-L1) pathway to be a key player in evading host immune responses. The strategy of immune checkpoint blockade is to reduce inhibitory signaling and restore the patient's natural tumor-specific T-cell-mediated immune responses. Nivolumab is the first PD-1 inhibitor to have gained approval for the treatment of patients with metastatic melanoma, squamous and nonsquamous non-small cell lung cancer (NSCLC), Hodgkin disease and recently RCC. In this review, we discuss results from studies of nivolumab in RCC, clinical experience with this agent, and its future development.
Collapse
Affiliation(s)
- Camille Mazza
- Department of Medical Oncology, Gustave Roussy, Villejuif, France
| | - Bernard Escudier
- Département de médecine Oncologique, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Laurence Albiges
- Département de médecine Oncologique, Gustave Roussy, Université Paris-Saclay, Villejuif, F-94805, 114 Rue Edouard Vaillant, 94800 Villejuif, France
| |
Collapse
|
24
|
Abstract
Anticancer immunotherapy has undergone a long evolving journey for decades, and has been dramatically applied to mainstream treatments in oncology in recent 5 years. This progress represents an advanced milestone following cytotoxic medicine and targeted therapy. Cellular immunity plays a pivotal role in the immune responses of hosts to tumor antigens. Such immunity is notably suppressed during neoplastic progression due to immuno-editing processes. Cellular immunity can also be selectively re-activated to combat malignancies while exploiting the advantages of contemporary scientific breakthroughs in molecular immunology and genetic engineering. The rapid advancement of cellular immunity-based therapeutic approaches has achieved high efficacy in certain cancer patients. Consequently, the landscape of oncologic medicine and pharmaceutical innovation has transformed recently. In this regard, we present a comprehensive update on clinically established anti-cancer treatments with cell immunity augmentation as the major mechanism of action.
Collapse
Affiliation(s)
- Daohong Chen
- Research Institute of Biomedicine, Yiling Pharmacy, Shijiazhuang 050035, China
| | - Xiaoshi Zhang
- Research Institute of Biomedicine, Yiling Pharmacy, Shijiazhuang 050035, China
| |
Collapse
|
25
|
Savoia P, Astrua C, Fava P. Ipilimumab (Anti-Ctla-4 Mab) in the treatment of metastatic melanoma: Effectiveness and toxicity management. Hum Vaccin Immunother 2016; 12:1092-101. [PMID: 26889818 PMCID: PMC4963052 DOI: 10.1080/21645515.2015.1129478] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 11/19/2015] [Accepted: 12/04/2015] [Indexed: 12/25/2022] Open
Abstract
In the last years the onset of new therapies changed the management of malignant melanoma. Anti CTLA-4 antibody ipilimumab was the first drug to achieve a significant improvement in survival of advanced stage melanoma. This new therapeutic agent is characterized by a number of side effects that are totally different from those of traditional chemotherapy, mainly caused by the immune system activation. The purpose of this paper is to underline the central role of ipilimumab in the treatment of metastatic melanoma and to characterize related adverse events in terms of incidence, duration and severity of presentation. The early recognition of these side effects is crucial in order to ensure an appropriate management of the toxicities, thus reducing the long term clinical sequelae and the inappropriate treatment discontinuation.
Collapse
Affiliation(s)
- Paola Savoia
- Department of Medical Sciences, University of Turin, Turin, Italy
- Department of Health Science, “A. Avogadro” University of Eastern Piedmont, Novara, Italy
| | - Chiara Astrua
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Paolo Fava
- Department of Medical Sciences, University of Turin, Turin, Italy
| |
Collapse
|
26
|
Zhu X, Lang J. The significance and therapeutic potential of PD-1 and its ligands in ovarian cancer: A systematic review. Gynecol Oncol 2016; 142:184-189. [PMID: 27063803 DOI: 10.1016/j.ygyno.2016.04.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 03/28/2016] [Accepted: 04/01/2016] [Indexed: 02/07/2023]
Abstract
Surgery, radiotherapy and chemotherapy are the mainstay of malignant cancer treatments. However, with the development of immunology, the emerging immunotherapy represents a rational and alternative approach for the treatment of human cancer, including ovarian cancer (OC). Based on a body of evidence and the clinical success of immunotherapy in many malignancies, it is confirmed that blocking the programmed death 1 (PD-1) and its ligands in OC is feasible and valid both in animal models and patients. Immunotherapy may play a significant role in the future clinical management and improve the prognosis of OC. This review will focus on the biological functions, treatment response, toxicity and viable target of PD-1 and its ligands in OC. Recognition of the multiple functions of PD-1 and its ligands in ovarian cancer will serve to deepen our understanding of the nature of OC, develop novel immunotherapy approaches and discover possible diagnostic and prognostic biomarkers in future clinical decisions.
Collapse
Affiliation(s)
- Xinxin Zhu
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Jinghe Lang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China.
| |
Collapse
|
27
|
Nouri N, Garbe C. Intralesional immunotherapy as a strategy to treat melanoma. Expert Opin Biol Ther 2016; 16:619-26. [DOI: 10.1517/14712598.2016.1157161] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
28
|
Daniele B, Sangro B, Petrylak D, Calabrò F, Cartenì G, Montesarchio V, De Placido S, Ascierto PA. The 2nd meeting of the Campania Society of Oncology Immunotherapy (SCITO): focus on hepatocellular carcinoma, kidney and bladder cancer. J Immunother Cancer 2016. [PMCID: PMC4717673 DOI: 10.1186/s40425-015-0105-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
In this paper, we review recent advances in immuno-oncology with particular focus on liver, kidney, and bladder cancers as discussed at the 2nd meeting of the Campania Society of Oncology Immunotherapy (SCITO).
Collapse
|
29
|
Abascal MF, Besso MJ, Rosso M, Mencucci MV, Aparicio E, Szapiro G, Furlong LI, Vazquez-Levin MH. CDH1/E-cadherin and solid tumors. An updated gene-disease association analysis using bioinformatics tools. Comput Biol Chem 2015; 60:9-20. [PMID: 26674224 DOI: 10.1016/j.compbiolchem.2015.10.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2015] [Revised: 10/17/2015] [Accepted: 10/19/2015] [Indexed: 12/13/2022]
Abstract
Cancer is a group of diseases that causes millions of deaths worldwide. Among cancers, Solid Tumors (ST) stand-out due to their high incidence and mortality rates. Disruption of cell-cell adhesion is highly relevant during tumor progression. Epithelial-cadherin (protein: E-cadherin, gene: CDH1) is a key molecule in cell-cell adhesion and an abnormal expression or/and function(s) contributes to tumor progression and is altered in ST. A systematic study was carried out to gather and summarize current knowledge on CDH1/E-cadherin and ST using bioinformatics resources. The DisGeNET database was exploited to survey CDH1-associated diseases. Reported mutations in specific ST were obtained by interrogating COSMIC and IntOGen tools. CDH1 Single Nucleotide Polymorphisms (SNP) were retrieved from the dbSNP database. DisGeNET analysis identified 609 genes annotated to ST, among which CDH1 was listed. Using CDH1 as query term, 26 disease concepts were found, 21 of which were neoplasms-related terms. Using DisGeNET ALL Databases, 172 disease concepts were identified. Of those, 80 ST disease-related terms were subjected to manual curation and 75/80 (93.75%) associations were validated. On selected ST, 489 CDH1 somatic mutations were listed in COSMIC and IntOGen databases. Breast neoplasms had the highest CDH1-mutation rate. CDH1 was positioned among the 20 genes with highest mutation frequency and was confirmed as driver gene in breast cancer. Over 14,000 SNP for CDH1 were found in the dbSNP database. This report used DisGeNET to gather/compile current knowledge on gene-disease association for CDH1/E-cadherin and ST; data curation expanded the number of terms that relate them. An updated list of CDH1 somatic mutations was obtained with COSMIC and IntOGen databases and of SNP from dbSNP. This information can be used to further understand the role of CDH1/E-cadherin in health and disease.
Collapse
Affiliation(s)
- María Florencia Abascal
- Laboratory of Cell-Cell Interaction in Cancer and Reproduction, Instituto de Biología & Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Fundación IBYME (FIBYME), Vuelta de Obligado 2490, Zip Code C1428ADN, Buenos Aires, Argentina.
| | - María José Besso
- Laboratory of Cell-Cell Interaction in Cancer and Reproduction, Instituto de Biología & Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Fundación IBYME (FIBYME), Vuelta de Obligado 2490, Zip Code C1428ADN, Buenos Aires, Argentina.
| | - Marina Rosso
- Laboratory of Cell-Cell Interaction in Cancer and Reproduction, Instituto de Biología & Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Fundación IBYME (FIBYME), Vuelta de Obligado 2490, Zip Code C1428ADN, Buenos Aires, Argentina.
| | - María Victoria Mencucci
- Laboratory of Cell-Cell Interaction in Cancer and Reproduction, Instituto de Biología & Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Fundación IBYME (FIBYME), Vuelta de Obligado 2490, Zip Code C1428ADN, Buenos Aires, Argentina.
| | - Evangelina Aparicio
- Laboratory of Cell-Cell Interaction in Cancer and Reproduction, Instituto de Biología & Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Fundación IBYME (FIBYME), Vuelta de Obligado 2490, Zip Code C1428ADN, Buenos Aires, Argentina.
| | - Gala Szapiro
- Laboratory of Cell-Cell Interaction in Cancer and Reproduction, Instituto de Biología & Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Fundación IBYME (FIBYME), Vuelta de Obligado 2490, Zip Code C1428ADN, Buenos Aires, Argentina.
| | - Laura Inés Furlong
- Research Programme on Biomedical Informatics (GRIB) (IMIM), DCEXS, Universitat Pompeu Fabra, C/Dr Aiguader 88, Zip Code 08003, Barcelona, Spain.
| | - Mónica Hebe Vazquez-Levin
- Laboratory of Cell-Cell Interaction in Cancer and Reproduction, Instituto de Biología & Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Fundación IBYME (FIBYME), Vuelta de Obligado 2490, Zip Code C1428ADN, Buenos Aires, Argentina; Laboratory of Cell-Cell Interaction in Cancer and Reproduction, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Fundación IBYME (FIBYME), Vuelta de Obligado 2490, Zip Code C1428ADN, Buenos Aires, Argentina.
| |
Collapse
|
30
|
Waugh KA, Leach SM, Slansky JE. Targeting Transcriptional Regulators of CD8+ T Cell Dysfunction to Boost Anti-Tumor Immunity. Vaccines (Basel) 2015; 3:771-802. [PMID: 26393659 PMCID: PMC4586477 DOI: 10.3390/vaccines3030771] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 09/09/2015] [Accepted: 09/10/2015] [Indexed: 02/07/2023] Open
Abstract
Transcription is a dynamic process influenced by the cellular environment: healthy, transformed, and otherwise. Genome-wide mRNA expression profiles reflect the collective impact of pathways modulating cell function under different conditions. In this review we focus on the transcriptional pathways that control tumor infiltrating CD8+ T cell (TIL) function. Simultaneous restraint of overlapping inhibitory pathways may confer TIL resistance to multiple mechanisms of suppression traditionally referred to as exhaustion, tolerance, or anergy. Although decades of work have laid a solid foundation of altered transcriptional networks underlying various subsets of hypofunctional or “dysfunctional” CD8+ T cells, an understanding of the relevance in TIL has just begun. With recent technological advances, it is now feasible to further elucidate and utilize these pathways in immunotherapy platforms that seek to increase TIL function.
Collapse
Affiliation(s)
- Katherine A Waugh
- University of Colorado School of Medicine, 12800 East 19th Avenue, Mail Stop 8333, Aurora, CO 80045, USA.
| | - Sonia M Leach
- Center for Genes, Environment and Health, National Jewish Health, Denver, CO 80206, USA.
| | - Jill E Slansky
- University of Colorado School of Medicine, 12800 East 19th Avenue, Mail Stop 8333, Aurora, CO 80045, USA.
| |
Collapse
|
31
|
Vela M, Aris M, Llorente M, Garcia-Sanz JA, Kremer L. Chemokine receptor-specific antibodies in cancer immunotherapy: achievements and challenges. Front Immunol 2015; 6:12. [PMID: 25688243 PMCID: PMC4311683 DOI: 10.3389/fimmu.2015.00012] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 01/07/2015] [Indexed: 12/22/2022] Open
Abstract
The 1990s brought a burst of information regarding the structure, expression pattern, and role in leukocyte migration and adhesion of chemokines and their receptors. At that time, the FDA approved the first therapeutic antibodies for cancer treatment. A few years later, it was reported that the chemokine receptors CXCR4 and CCR7 were involved on directing metastases to liver, lung, bone marrow, or lymph nodes, and the over-expression of CCR4, CCR6, and CCR9 by certain tumors. The possibility of inhibiting the interaction of chemokine receptors present on the surface of tumor cells with their ligands emerged as a new therapeutic approach. Therefore, many research groups and companies began to develop small molecule antagonists and specific antibodies, aiming to neutralize signaling from these receptors. Despite great expectations, so far, only one anti-chemokine receptor antibody has been approved for its clinical use, mogamulizumab, an anti-CCR4 antibody, granted in Japan to treat refractory adult T-cell leukemia and lymphoma. Here, we review the main achievements obtained with anti-chemokine receptor antibodies for cancer immunotherapy, including discovery and clinical studies, proposed mechanisms of action, and therapeutic applications.
Collapse
Affiliation(s)
- Maria Vela
- Department of Immunology and Oncology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB/CSIC), Madrid, Spain
| | - Mariana Aris
- Centro de Investigaciones Oncológicas, Fundación Cáncer, Buenos Aires, Argentina
| | - Mercedes Llorente
- Protein Tools Unit, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB/CSIC), Madrid, Spain
| | - Jose A. Garcia-Sanz
- Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CIB/CSIC), Madrid, Spain
| | - Leonor Kremer
- Department of Immunology and Oncology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB/CSIC), Madrid, Spain
- Protein Tools Unit, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB/CSIC), Madrid, Spain
| |
Collapse
|
32
|
|