101
|
Rijavec E, Genova C, Biello F, Rossi G, Indini A, Grossi F. Current state of the art and future perspectives with immunotherapy in the management of small cell lung cancer. Expert Rev Respir Med 2021; 15:1427-1435. [PMID: 34590937 DOI: 10.1080/17476348.2021.1987887] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Small cell lung cancer (SCLC) is an aggressive tumor with a severe prognosis. At the time of diagnosis, most patients present with extensive-stage (ES) disease. For decades, platinum-based chemotherapy has been the only pillar of SCLC treatment, but now, the clinical management of this disease is rapidly evolving thanks to the introduction of immune checkpoint inhibitors (ICIs). AREAS COVERED In this review, we describe the most recent advances in the treatment of SCLC and discuss the emerging challenges associated with ICI treatments. Meaningful data were collected from the currently available literature on PubMed and in international oncology meetings. EXPERT OPINION Recently, meaningful improvements in outcomes of SCLC patients have been achieved with anti-PD-L1 atezolizumab or durvalumab combined with chemotherapy in first line. Results of studies evaluating the role of ICIs in limited-stage (LS) SCLC patients are awaited. Further efforts are required to better understand the role of immunotherapy in the treatment of SCLC and to identify patients most likely to benefit from this treatment strategy.
Collapse
Affiliation(s)
- Erika Rijavec
- Medical Oncology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Carlo Genova
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Federica Biello
- Division of Oncology, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Giovanni Rossi
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy.,Medical Oncology Unit, Ospedale Padre Antero Micone, Genova, Italy
| | - Alice Indini
- Medical Oncology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Francesco Grossi
- Division of Medical Oncology, University of Insubria, Asst Dei Sette Laghi, Varese, Italy
| |
Collapse
|
102
|
Hiddinga BI, Raskin J, Janssens A, Pauwels P, Van Meerbeeck JP. Recent developments in the treatment of small cell lung cancer. Eur Respir Rev 2021; 30:210079. [PMID: 34261744 PMCID: PMC9488550 DOI: 10.1183/16000617.0079-2021] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 04/27/2021] [Indexed: 12/19/2022] Open
Abstract
Small cell lung cancer (SCLC) comprises about 15% of all lung cancers. It is an aggressive disease, with early metastasis and a poor prognosis. Until recently, SCLC treatment remained relatively unchanged, with chemotherapy remaining the cornerstone of treatment. In this overview we will highlight the recent advances in the field of staging, surgery, radiotherapy and systemic treatment. Nevertheless, the prognosis remains dismal and there is a pressing need for new treatment options. We describe the progress that has been made in systemic treatment by repurposing existing drugs and the addition of targeted treatment. In recent years, immunotherapy entered the clinic with high expectations of its role in the treatment of SCLC. Unravelling of the genomic sequence revealed new possible targets that may act as biomarkers in future treatment of patients with SCLC. Hopefully, in the near future, we will be able to identify patients who may benefit from targeted therapy or immunotherapy to improve prognoses.
Collapse
Affiliation(s)
- Birgitta I Hiddinga
- Dept of Pulmonary Medicine and Tuberculosis, University Medical Centre Groningen, Groningen, The Netherlands
- Both authors contributed equally
| | - Jo Raskin
- Dept of Thoracic Oncology, Antwerp University Hospital, Edegem, Belgium
- Both authors contributed equally
| | - Annelies Janssens
- Dept of Thoracic Oncology, Antwerp University Hospital, Edegem, Belgium
- University of Antwerp, Antwerp, Belgium
| | - Patrick Pauwels
- University of Antwerp, Antwerp, Belgium
- Dept of Pathology, Antwerp University Hospital, Edegem, Belgium
- European Reference Network for rare and low prevalent lung diseases (ERN-LUNG), Frankfurt am Main, Germany
| | - Jan P Van Meerbeeck
- Dept of Thoracic Oncology, Antwerp University Hospital, Edegem, Belgium
- University of Antwerp, Antwerp, Belgium
- European Reference Network for rare and low prevalent lung diseases (ERN-LUNG), Frankfurt am Main, Germany
| |
Collapse
|
103
|
Lee S, Shim HS, Ahn BC, Lim SM, Kim HR, Cho BC, Hong MH. Efficacy and safety of atezolizumab, in combination with etoposide and carboplatin regimen, in the first-line treatment of extensive-stage small-cell lung cancer: a single-center experience. Cancer Immunol Immunother 2021; 71:1093-1101. [PMID: 34568975 DOI: 10.1007/s00262-021-03052-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 09/06/2021] [Indexed: 01/10/2023]
Abstract
BACKGROUND Patients with small-cell lung cancer (SCLC) have a dismal prognosis with limited overall survival (OS) despite a high response rate to chemotherapy. Recently, immune checkpoint inhibitors, combined with chemotherapy, as the first-line treatment for extensive-stage (ES)-SCLC have shown improvement in clinical outcomes. PATIENTS AND METHODS Real-world data from 68 Korean ES-SCLC patients, treated with atezolizumab, etoposide, and carboplatin at Yonsei Cancer Center between June 2019 and November 2020, were retrospectively analyzed to determine safety and efficacy using Cox regression analysis. RESULTS The median follow-up was 11.6 months. The median progression-free survival was 4.6 months (95% confidence interval [CI] 4.0-5.2), and the median OS was 12.0 months (95% CI 7.4-16.6). Baseline bone metastasis, immune-related adverse events (IRAEs), and elevated LDH were related to OS (hazard ratio 2.18, 0.33, and 4.64; P = 0.05, 0.02, and 0.003, respectively). Among the 42 patients with disease progression, liver metastasis progression and baseline bone metastasis were associated with inferior OS, but without statistical significance (hazard ratio 2.47 and 1.97; P = 0.25 and 0.26, respectively). Overall, 61 (89.7%) patients experienced treatment-related adverse events (TRAEs), with hematologic toxicities as the most common grade 3-4 TRAEs. Twenty-two (32.4%) patients experienced IRAEs, with skin rash as the most common, and five (7.4%) patients had grade-3 IRAEs (pneumonitis, hyperglycemia, and aspartate aminotransferase elevation). CONCLUSION Atezolizumab, combined with etoposide and carboplatin, showed efficacy and safety in our real-world data. Further studies are needed to predict the response to immunotherapy in SCLC.
Collapse
Affiliation(s)
- Seoyoung Lee
- Division of Medical Oncology, Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Hyo Sup Shim
- Department of Pathology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - Beung-Chul Ahn
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Severance Hospital, Yonsei University College of Medicine, Yonsei-ro 50-1, Seodaemun-gu, Seoul, 03722, Korea
| | - Sun Min Lim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Severance Hospital, Yonsei University College of Medicine, Yonsei-ro 50-1, Seodaemun-gu, Seoul, 03722, Korea
| | - Hye Ryun Kim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Severance Hospital, Yonsei University College of Medicine, Yonsei-ro 50-1, Seodaemun-gu, Seoul, 03722, Korea
| | - Byoung Chul Cho
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Severance Hospital, Yonsei University College of Medicine, Yonsei-ro 50-1, Seodaemun-gu, Seoul, 03722, Korea
| | - Min Hee Hong
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Severance Hospital, Yonsei University College of Medicine, Yonsei-ro 50-1, Seodaemun-gu, Seoul, 03722, Korea.
| |
Collapse
|
104
|
Peters S, Pujol JL, Dafni U, Dómine M, Popat S, Reck M, Andrade J, Becker A, Moro-Sibilot D, Curioni-Fontecedro A, Molinier O, Nackaerts K, Insa Mollá A, Gervais R, López Vivanco G, Madelaine J, Mazieres J, Faehling M, Griesinger F, Majem M, González Larriba JL, Provencio Pulla M, Vervita K, Roschitzki-Voser H, Ruepp B, Mitchell P, Stahel RA, Le Pechoux C, De Ruysscher D. Consolidation nivolumab and ipilimumab versus observation in limited-disease small-cell lung cancer after chemo-radiotherapy - results from the randomised phase II ETOP/IFCT 4-12 STIMULI trial. Ann Oncol 2021; 33:67-79. [PMID: 34562610 DOI: 10.1016/j.annonc.2021.09.011] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 08/13/2021] [Accepted: 09/12/2021] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Concurrent chemotherapy and thoracic radiotherapy followed by prophylactic cranial irradiation (PCI) is the standard treatment in limited-disease small-cell lung cancer (LD-SCLC), with 5-year overall survival (OS) of only 25% to 33%. PATIENTS AND METHODS STIMULI is a 1:1 randomised phase II trial aiming to demonstrate superiority of consolidation combination immunotherapy versus observation after chemo-radiotherapy plus PCI (protocol amendment-1). Consolidation immunotherapy consisted of four cycles of nivolumab [1 mg/kg, every three weeks (Q3W)] plus ipilimumab (3 mg/kg, Q3W), followed by nivolumab monotherapy (240 mg, Q2W) for up to 12 months. Patient recruitment closed prematurely due to slow accrual and the statistical analyses plan was updated to address progression-free survival (PFS) as the only primary endpoint. RESULTS Of the 222 patients enrolled, 153 were randomised (78: experimental; 75: observation). Among the randomised patients, median age was 62 years, 60% males, 34%/65% current/former smokers, 31%/66% performance status (PS) 0/1. Up to 25 May 2020 (median follow-up 22.4 months), 40 PFS events were observed in the experimental arm, with median PFS 10.7 months [95% confidence interval (CI) 7.0-not estimable (NE)] versus 42 events and median 14.5 months (8.2-NE) in the observation, hazard ratio (HR) = 1.02 (0.66-1.58), two-sided P = 0.93. With updated follow-up (03 June 2021; median: 35 months), median OS was not reached in the experimental arm, while it was 32.1 months (26.1-NE) in observation, with HR = 0.95 (0.59-1.52), P = 0.82. In the experimental arm, median time-to-treatment-discontinuation was only 1.7 months. CTCAE v4 grade ≥3 adverse events were experienced by 62% of patients in the experimental and 25% in the observation arm, with 4 and 1 fatal, respectively. CONCLUSIONS The STIMULI trial did not meet its primary endpoint of improving PFS with nivolumab-ipilimumab consolidation after chemo-radiotherapy in LD-SCLC. A short period on active treatment related to toxicity and treatment discontinuation likely affected the efficacy results.
Collapse
Affiliation(s)
- S Peters
- Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland; Lausanne University, Lausanne, Switzerland
| | - J-L Pujol
- Thoracic Oncology Unit, Hopital Arnaud de Villeneuve, Montpellier, France
| | - U Dafni
- National and Kapodistrian University of Athens, Athens, Greece; Frontier Science Foundation-Hellas, Athens, Greece
| | - M Dómine
- Hospital Universitario Fundacion Jimenez Díaz (IIS-FJD), Madrid, Spain
| | - S Popat
- Medicine, Royal Marsden Hospital, London, UK
| | - M Reck
- Thoracic Oncology, Lung Clinic Grosshansdorf, Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Grosshansdorf, Germany
| | - J Andrade
- Medical Oncology, Hospital Virgen De La Salud, Toledo, Spain
| | - A Becker
- Department of Pulmonology, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - D Moro-Sibilot
- Thoracic Oncology Unit, Centre Hospitalier Universitaire Grenoble-Alpes, Grenoble, France
| | - A Curioni-Fontecedro
- Department of Medical Oncology and Hematology, University Hospital Zürich, Zürich, Switzerland
| | - O Molinier
- Department of Respiratory Disease, Centre Hospitalier - Le Mans, Le Mans, France
| | - K Nackaerts
- Department of Pulmonology, Respiratory Oncology Unit, KU Leuven, Leuven, Belgium
| | - A Insa Mollá
- Medical Oncology, Hospital Clínico Universitario De Valencia, Valencia, Spain
| | - R Gervais
- Medical Oncology, Centre François Baclesse, Caen, France
| | - G López Vivanco
- Medical Oncology, Hospital Universitario Cruces, Barakaldo, Spain
| | - J Madelaine
- Thoracic Oncology Unit, Centre Hospitalier Universitaire Caen Normandie, Caen, France
| | - J Mazieres
- Thoracic Oncology, Centre Hospitalier Universitaire Toulouse, Toulouse, France
| | | | | | - M Majem
- Hospital De La Santa Creu I Sant Pau, Barcelona, Spain
| | | | | | - K Vervita
- Frontier Science Foundation-Hellas, Athens, Greece
| | - H Roschitzki-Voser
- Coordinating Office, European Thoracic Oncology Platform, Bern, Switzerland
| | - B Ruepp
- Coordinating Office, European Thoracic Oncology Platform, Bern, Switzerland
| | - P Mitchell
- Olivia Newton-John Cancer Centre, Austin Hospital (VIC), Melbourne, Australia
| | - R A Stahel
- Coordinating Office, European Thoracic Oncology Platform, Bern, Switzerland.
| | - C Le Pechoux
- Department of Radiation Oncology, Gustave Roussy, Villejuif, France
| | - D De Ruysscher
- Maastricht University Medical Center, Department of Radiation Oncology (Maastro), GROW School for Oncology, Maastricht, Netherlands
| | | |
Collapse
|
105
|
Hao Z, Sekkath Veedu J. Current Strategies for Extensive Stage Small Cell Lung Cancer Beyond First-line Therapy. Clin Lung Cancer 2021; 23:14-20. [PMID: 34656433 DOI: 10.1016/j.cllc.2021.09.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 08/22/2021] [Accepted: 09/09/2021] [Indexed: 02/07/2023]
Abstract
Extensive stage small cell lung cancer carries extremely poor prognosis and adding immune checkpoint inhibitor to platinum etoposide combination in first line only improved outcomes modestly. Once disease recurs, treatment response is only transient in nature. Various strategies that are being explored include dual checkpoint blockade, BiTE and CAR-T cell approaches. Immune checkpoint inhibitors are being combined with PARP inhibitors. Other approaches currently being investigated include liposomal irinotecan and combining known active agents for SCLC in relapsed setting such as newly approved lurbinectedin with doxorubicin, paclitaxel, irinotecan or topotecan with ATR inhibitor (Berzosertib). Temozolomide has also been tested in combination with a Parp inhibitor. New antibody or small molecule drug conjugates are being actively investigated, so is a biomarker based approach. Better understanding of small cell lung cancer disease biology via high through-put genomic, proteomic and methylation profiling offer glimpse of hope in our efforts to contain this deadly disease. A table of representative molecular targets under investigation is provided in the end.
Collapse
Affiliation(s)
- Zhonglin Hao
- Division of Medical Oncology, Department of Medicine, Markey Cancer Center, College of Medicine, University of Kentucky, Lexington KY.
| | - Janeesh Sekkath Veedu
- Division of Medical Oncology, Department of Medicine, Markey Cancer Center, College of Medicine, University of Kentucky, Lexington KY
| |
Collapse
|
106
|
He M, Wu B, Liu Q, Fang Z, Liu M, Yi F, Wei Y, Peng J, Zhang W. Topotecan plus Platinum-Based Chemotherapy versus Etoposide plus Platinum-Based Chemotherapy for Small-Cell Lung Cancer: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Chemotherapy 2021; 66:113-123. [PMID: 34515066 DOI: 10.1159/000517990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 06/19/2021] [Indexed: 11/19/2022]
Abstract
BACKGROUND Whether topotecan plus platinum-based chemotherapy (TP) can achieve better results than etoposide plus platinum-based chemotherapy (EP) for small-cell lung cancer (SCLC) treatment is still controversial in clinical applications. We compared the effectiveness and toxicity of TP versus EP in this meta-analysis. METHODS We searched PubMed, ScienceDirect, Cochrane Library, Scopus, Ovid MEDLINE, Embase, Web of Science, and Google Scholar databases for completeness one by one to find articles that met the conditions. Overall survival (OS) and progression-free survival (PFS) were analyzed as primary endpoints, and the objective response rate (ORR), disease control rate (DCR), and adverse events (AEs) were analyzed as secondary endpoints. RESULTS In total, 2,480 articles were retrieved, and 6 randomized controlled trials (RCTs) contained results based on 1,924 patients. EP suggested conspicuously better OS (hazard ratio [HR]: 1.24 [1.02, 1.50], p = 0.03) and PFS (HR: 1.39 [1.17, 1.64], p = 0.0001) in SCLC treatment than TP, and ORR (54.1% vs. 60.2%, risk ratio [RR]: 0.77 [0.57, 1.06], p = 0.11), and DCR (74.9% vs. 84.4%, RR: 0.89 [0.79, 1.00], p = 0.06) tended to favor EP. Subgroup analysis of subsistence showed that EP had prominent benefit in the following subgroups: Asian, median age > 60, first-line treatment, ECOG 0-2, intravenous topotecan, and cisplatin. AEs illustrated that EP had conspicuously more anemia and alopecia than TP. CONCLUSIONS Compared with TP, EP was noticeably better in OS and PFS, but EP was toxic in terms of anemia and alopecia. More multicenter, better planned RCTs are needed.
Collapse
Affiliation(s)
- Mengyu He
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Bingxuan Wu
- Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Qiangyun Liu
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Zige Fang
- Jiangxi Medical College, Nanchang University, Nanchang, China.,Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Miaowen Liu
- Jiangxi Medical College, Nanchang University, Nanchang, China.,Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Fengming Yi
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yiping Wei
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jinhua Peng
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Wenxiong Zhang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
107
|
Albertelli M, Dotto A, Nista F, Veresani A, Patti L, Gay S, Sciallero S, Boschetti M, Ferone D. "Present and future of immunotherapy in Neuroendocrine Tumors". Rev Endocr Metab Disord 2021; 22:615-636. [PMID: 33851319 PMCID: PMC8346388 DOI: 10.1007/s11154-021-09647-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/16/2021] [Indexed: 12/13/2022]
Abstract
Immunotherapy, so promising in many neoplasms, still does not have a precise role in the treatment of neuroendocrine neoplasms (NENs). In this article, we provide an overview on the current knowledge about immunotherapy with immune checkpoint inhibitors (ICIs) applied to NENs, evaluating future perspectives in this setting of tumors.Evidence so far available for ICIs in gastroenteropancreatic (GEP)-NENs is definitively not as robust as for other tumors such as Small Cell Lung Cancer or Merkel Cell Carcinoma. In fact, with regard to the well-differentiated forms of NENs (NETs), the results obtained nowadays have been disappointing. However, the near future, might reserve interesting results for ICIs in GEP-NEN from a total of nine different ICI drugs, used throughout 19 randomised controlled trials. Such numbers highlight the growing attention gathering around NENs and ICIs, in response to the need of stronger evidences supporting such therapy.For the future, the most important aspect will be to study strategies that can make NETs more susceptible to response to ICI and, thus, enhance the effectiveness of these treatments. Therefore, the combination of conventional therapy, target therapy and immunotherapy deserve attention and warrant to be explored. A sequential chemotherapy, possibly inducing an increase in tumor mutational burden and tested before immunotherapy, could be a hypothesis deserving more consideration. A radiation treatment that increases tumor-infiltrating lymphocytes, could be another approach to explore before ICIs in NENs. Equally essential will be the identification of biomarkers useful for selecting patients potentially responsive to this type of treatment.
Collapse
Affiliation(s)
- Manuela Albertelli
- Endocrinology Unit, IRCCS AOU San Martino, Genoa, Italy.
- Endocrinology Unit, Department of Internal Medicine and Medical Specialties (DiMI), Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy.
| | - Andrea Dotto
- Endocrinology Unit, Department of Internal Medicine and Medical Specialties (DiMI), Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy
| | - Federica Nista
- Endocrinology Unit, Department of Internal Medicine and Medical Specialties (DiMI), Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy
| | - Alessandro Veresani
- Endocrinology Unit, Department of Internal Medicine and Medical Specialties (DiMI), Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy
| | - Luca Patti
- Endocrinology Unit, Department of Internal Medicine and Medical Specialties (DiMI), Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy
| | - Stefano Gay
- Endocrinology Unit, Department of Internal Medicine and Medical Specialties (DiMI), Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy
| | | | - Mara Boschetti
- Endocrinology Unit, IRCCS AOU San Martino, Genoa, Italy
- Endocrinology Unit, Department of Internal Medicine and Medical Specialties (DiMI), Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy
| | - Diego Ferone
- Endocrinology Unit, IRCCS AOU San Martino, Genoa, Italy
- Endocrinology Unit, Department of Internal Medicine and Medical Specialties (DiMI), Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy
| |
Collapse
|
108
|
Blackhall F, Jao K, Greillier L, Cho BC, Penkov K, Reguart N, Majem M, Nackaerts K, Syrigos K, Hansen K, Schuette W, Cetnar J, Cappuzzo F, Okamoto I, Erman M, Langer SW, Kato T, Groen H, Sun Z, Luo Y, Tanwani P, Caffrey L, Komarnitsky P, Reinmuth N. Efficacy and Safety of Rovalpituzumab Tesirine Compared With Topotecan as Second-Line Therapy in DLL3-High SCLC: Results From the Phase 3 TAHOE Study. J Thorac Oncol 2021; 16:1547-1558. [PMID: 33607312 DOI: 10.1016/j.jtho.2021.02.009] [Citation(s) in RCA: 167] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/29/2021] [Accepted: 02/09/2021] [Indexed: 12/12/2022]
Abstract
INTRODUCTION DLL3, an atypical Notch ligand, is expressed in SCLC tumors but is not detectable in normal adult tissues. Rovalpituzumab tesirine (Rova-T) is an antibody-drug conjugate containing a DLL3-targeting antibody tethered to a cytotoxic agent pyrrolobenzodiazepine by means of a protease-cleavable linker. The efficacy and safety of Rova-T compared with topotecan as second-line therapy in patients with SCLC expressing high levels of DLL3 (DLL3-high) was evaluated. METHODS The TAHOE study was an open-label, two-to-one randomized, phase 3 study comparing Rova-T with topotecan as second-line therapy in DLL3-high advanced or metastatic SCLC. Rova-T (0.3 mg/kg) was administered intravenously on day 1 of a 42-day cycle for two cycles, with two additional cycles available to patients who met protocol-defined criteria for continued dosing. Topotecan (1.5 mg/m2) was administered intravenously on days 1 to 5 of a 21-day cycle. The primary end point was overall survival (OS). RESULTS Patients randomized to Rova-T (n = 296) and topotecan (n = 148) were included in the efficacy analyses. The median age was 64 years, and 77% had the extensive disease at initial diagnosis. The median OS (95% confidence interval) was 6.3 months (5.6-7.3) in the Rova-T arm and 8.6 months (7.7-10.1) in the topotecan arm (hazard ratio, 1.46 [95% confidence interval: 1.17-1.82]). An independent data monitoring committee recommended that enrollment be discontinued because of the shorter OS observed with Rova-T compared with topotecan. Safety profiles for both drugs were consistent with previous reports. CONCLUSIONS Compared with topotecan, which is the current standard second-line chemotherapy, Rova-T exhibited an inferior OS and higher rates of serosal effusions, photosensitivity reaction, and peripheral edema in patients with SCLC. A considerable unmet therapeutic need remains in this population.
Collapse
Affiliation(s)
- Fiona Blackhall
- Division of Cancer Sciences, The University of Manchester, Manchester, United Kingdom; Department of Medical Oncology, The Christie National Health Service (NHS) Foundation Trust, Manchester, United Kingdom.
| | - Kevin Jao
- Department of Hematology and Oncology, Hopital du Sacre Coeur Montreal, Montreal, Canada
| | - Laurent Greillier
- Multidisciplinary Oncology and Therapeutic Innovations Department, Centre de Recherche en Cancérologie de Marseille (CRCM), Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), Assistance Publique-Hopitaux de Marseille (APHM), Aix-Marseille University, Marseille, France
| | - Byoung Chul Cho
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea
| | | | - Noemi Reguart
- Department of Medical Oncology, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Hospital Clinic de Barcelona, Barcelona, Spain
| | - Margarita Majem
- Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Kristiaan Nackaerts
- Department of Pulmonology and Respiratory Oncology, University Hospital Leuven, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Konstantinos Syrigos
- Department of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Karin Hansen
- Department of Oncology, Odense Universitets Hospital, Odense, Denmark
| | - Wolfgang Schuette
- 2nd Medical Department, Krankenhaus Martha-Maria Halle-Doelau, Halle, Germany
| | - Jeremy Cetnar
- Department of Medicine, Oregon Health & Science University, Portland, Oregon
| | - Federico Cappuzzo
- Department of Medical Oncology, Istituto Nazionale Tumori Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Isamu Okamoto
- Department of Medical Oncology, Kyushu University Hospital, Fukuoka, Japan
| | - Mustafa Erman
- Department of Medical Oncology, Cancer Institute, Hacettepe University, Ankara, Turkey
| | - Seppo W Langer
- Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Terufumi Kato
- Department of Thoracic Oncology, Kanagawa Cancer Center, Yokohama, Japan
| | - Harry Groen
- Department of Pulmonary Disease, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | | | - Yan Luo
- AbbVie, Inc., North Chicago, Illinois
| | | | | | | | - Niels Reinmuth
- Thoracic Oncology Department, Asklepios Fachkliniken München-Gauting, Gauting, Germany
| |
Collapse
|
109
|
Remash D, Prince DS, McKenzie C, Strasser SI, Kao S, Liu K. Immune checkpoint inhibitor-related hepatotoxicity: A review. World J Gastroenterol 2021; 27:5376-5391. [PMID: 34539139 PMCID: PMC8409159 DOI: 10.3748/wjg.v27.i32.5376] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/28/2021] [Accepted: 08/03/2021] [Indexed: 02/06/2023] Open
Abstract
The application of immune checkpoint inhibitors (ICI) in advanced cancer has been a major development in the last decade. The indications for ICIs are constantly expanding into new territory across different cancers, disease stages and lines of therapy. With this increased use, adverse events including immune checkpoint inhibitor-related hepatotoxicity (ICH) have emerged as an important clinical problem. This along with the introduction of ICI as first- and second-line treatments for advanced hepatocellular carcinoma makes ICH very relevant to gastroenterologists and hepatologists. The incidence of ICH varies between 1%-20% depending on the number, type and dose of ICI received. Investigation and management generally involve excluding differential diagnoses and following a stepwise escalation of withholding or ceasing ICI, corticosteroid treatment and adding other immunosuppressive agents depending on the severity of toxicity. The majority of patients with ICH recover and some may even safely recommence ICI therapy. Guideline recommendations are largely based on evidence derived from retrospective case series which highlights a priority for future research.
Collapse
Affiliation(s)
- Devika Remash
- AW Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital, Sydney 2050, NSW, Australia
| | - David S Prince
- AW Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital, Sydney 2050, NSW, Australia
| | - Catriona McKenzie
- AW Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital, Sydney 2050, NSW, Australia
- Sydney Medical School, University of Sydney, Sydney 2006, NSW, Australia
- Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Sydney 2050, NSW, Australia
- New South Wales Health Pathology, New South Wales Health, Sydney 2050, NSW, Australia
| | - Simone I Strasser
- AW Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital, Sydney 2050, NSW, Australia
- Sydney Medical School, University of Sydney, Sydney 2006, NSW, Australia
| | - Steven Kao
- Sydney Medical School, University of Sydney, Sydney 2006, NSW, Australia
- Medical Oncology, Chris O’Brien Lifehouse, Sydney 2050, NSW, Australia
| | - Ken Liu
- AW Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital, Sydney 2050, NSW, Australia
- Sydney Medical School, University of Sydney, Sydney 2006, NSW, Australia
| |
Collapse
|
110
|
Wang DK, Zuo Q, He QY, Li B. Targeted Immunotherapies in Gastrointestinal Cancer: From Molecular Mechanisms to Implications. Front Immunol 2021; 12:705999. [PMID: 34447376 PMCID: PMC8383067 DOI: 10.3389/fimmu.2021.705999] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 07/26/2021] [Indexed: 12/12/2022] Open
Abstract
Gastrointestinal cancer is a leading cause of cancer-related mortality and remains a major challenge for cancer treatment. Despite the combined administration of modern surgical techniques and chemoradiotherapy (CRT), the overall 5-year survival rate of gastrointestinal cancer patients in advanced stage disease is less than 15%, due to rapid disease progression, metastasis, and CRT resistance. A better understanding of the mechanisms underlying cancer progression and optimized treatment strategies for gastrointestinal cancer are urgently needed. With increasing evidence highlighting the protective role of immune responses in cancer initiation and progression, immunotherapy has become a hot research topic in the integrative management of gastrointestinal cancer. Here, an overview of the molecular understanding of colorectal cancer, esophageal cancer and gastric cancer is provided. Subsequently, recently developed immunotherapy strategies, including immune checkpoint inhibitors, chimeric antigen receptor T cell therapies, tumor vaccines and therapies targeting other immune cells, have been described. Finally, the underlying mechanisms, fundamental research and clinical trials of each agent are discussed. Overall, this review summarizes recent advances and future directions for immunotherapy for patients with gastrointestinal malignancies.
Collapse
Affiliation(s)
| | | | | | - Bin Li
- Ministry of Education (MOE), Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, Jinan University, Guangzhou, China
| |
Collapse
|
111
|
Cheng Y, Wang Q, Li K, Shi J, Liu Y, Wu L, Han B, Chen G, He J, Wang J, Lou D, Yu H, Wang S, Qin H, Li X. Anlotinib vs placebo as third- or further-line treatment for patients with small cell lung cancer: a randomised, double-blind, placebo-controlled Phase 2 study. Br J Cancer 2021; 125:366-371. [PMID: 34006926 PMCID: PMC8329046 DOI: 10.1038/s41416-021-01356-3] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 02/19/2021] [Accepted: 03/11/2021] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND This study aimed to evaluate the efficacy and safety of anlotinib as a third-line and subsequent treatment for patients with small cell lung cancer (SCLC). METHODS We conducted this Phase 2 trial at 11 institutions in China. Patients with pathologically confirmed SCLC who failed at least two lines of chemotherapy were enrolled. Subjects were randomly assigned in a 2:1 ratio to receive either anlotinib 12 mg orally once daily for 14 days every 3 weeks or placebo. The primary endpoint was progression-free survival (PFS). RESULTS Between March 30, 2017 and June 8, 2018, a total of 82 and 38 patients were randomly assigned to receive anlotinib and placebo. The median PFS was significantly longer in the anlotinib group compared with the placebo group (4.1 months [95% confidence interval (CI), 2.8-4.2] vs 0.7 months [95% CI, 0.7-0.8]; hazard ratio (HR) 0.19 [95% CI, 0.12-0.32], p < 0.0001). Overall survival (OS) was significantly longer with anlotinib than placebo (7.3 months [95% CI, 6.1-10.3] vs 4.9 months [95% CI, 2.7-6.0]; HR 0.53 [95% CI, 0.34-0.81], p = 0.0029). CONCLUSIONS Anlotinib as a third-line or subsequent treatment for Chinese patients with SCLC showed improved PFS and OS than placebo with favourable safety profile. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov, number NCT03059797.
Collapse
Affiliation(s)
- Ying Cheng
- grid.440230.1Department of Thoracic Medical Oncology, Jilin Cancer Hospital, Changchun, China
| | - Qiming Wang
- grid.414008.90000 0004 1799 4638Department of Respiratory Medicine, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China ,grid.414008.90000 0004 1799 4638Department of Respiratory Medicine, Henan Cancer Hospital, Zhengzhou, China
| | - Kai Li
- grid.411918.40000 0004 1798 6427Department of Pulmonary Oncology, Tianjin Medical University Cancer Hospital, Tianjin, China
| | - Jianhua Shi
- Department of Medical Oncology, Linyi Cancer Hospital, Linyi, China
| | - Ying Liu
- grid.440230.1Department of Thoracic Medical Oncology, Jilin Cancer Hospital, Changchun, China
| | - Lin Wu
- grid.410622.30000 0004 1758 2377Department of Thoracic Medical Oncology, Hunan Cancer Hospital, Changsha, China
| | - Baohui Han
- grid.16821.3c0000 0004 0368 8293Department of Respiratory Medicine, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Gongyan Chen
- grid.412651.50000 0004 1808 3502Department of Respiratory Medicine, Harbin Medical University Cancer Hospital, Harbin, China
| | - Jianxing He
- grid.470124.4Department of Thoracic Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jie Wang
- grid.459409.50000 0004 0632 3230Department of Thoracic Medical Oncology, Cancer Hospital Chinese Academy of Medical Sciences, Beijing, China
| | - Donghua Lou
- grid.89957.3a0000 0000 9255 8984Department of Biostatistics, School of Public Health Nanjing Medical University, Nanjing, China
| | - Hao Yu
- grid.89957.3a0000 0000 9255 8984Department of Biostatistics, School of Public Health Nanjing Medical University, Nanjing, China
| | - Shanchun Wang
- Chia-tai Tianqing Pharmaceutical Group Co., Ltd, Nanjing, China
| | - Haifeng Qin
- grid.414252.40000 0004 1761 8894Department of Pulmonary Oncology, The Fifth Medical Centre of Chinese PLA General hospital, Beijing, China
| | - Xiaoling Li
- grid.459742.90000 0004 1798 5889Department of Medical Oncology, Liaoning Cancer Hospital and Institute, Shenyang, China
| |
Collapse
|
112
|
Abbas A, Gruner M, Karohl J, Rose PG, Joehlin-Price A, Stover D, Mahdi H. Case Report: Circulating Tumor DNA Fraction Analysis Using Ultra-Low-Pass Whole-Genome Sequencing Correlates Response to Chemoradiation and Recurrence in Stage IV Small-Cell Carcinoma of the Cervix - A Longitudinal Study. Front Oncol 2021; 11:652683. [PMID: 34381704 PMCID: PMC8350481 DOI: 10.3389/fonc.2021.652683] [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: 01/12/2021] [Accepted: 07/08/2021] [Indexed: 11/13/2022] Open
Abstract
Neuroendocrine carcinoma of the cervix is a rare and aggressive form of cervical cancer that presents with frequent metastasis at diagnosis and high recurrence rates. Primary treatment is multimodal, which often includes chemotherapy with or without radiation therapy. There are no data available to guide treatment for recurrence, and second-line therapies are extrapolated from small-cell lung carcinoma data. Close monitoring of these patients for recurrence is paramount. Evaluation of circulating tumor DNA (ctDNA) in the peripheral blood is an attractive approach due to its non-invasive nature. Ultra-low-pass whole-genome sequencing (ULP-WGS) can assess tumor burden and response to therapy and predict recurrence; however, data are lacking regarding the role of ULP-WGS in small-cell carcinoma of the cervix. This study demonstrates a patient whose response to chemotherapy and cancer recurrence was accurately monitored by ctDNA analysis using ULP-WGS and confirmed with radiologic imaging findings.
Collapse
Affiliation(s)
- Ata Abbas
- Division of Hematology and Oncology, Department of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Morgan Gruner
- Division of Gynecologic Oncology, Obstetrics, Gynecology and Women's Health Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Jennifer Karohl
- Division of Gynecologic Oncology, Obstetrics, Gynecology and Women's Health Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Peter G Rose
- Division of Gynecologic Oncology, Obstetrics, Gynecology and Women's Health Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Amy Joehlin-Price
- Department of Anatomic Pathology, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Daniel Stover
- Division of Medical Oncology, The Stefanie Spielman Comprehensive Breast Center, The James Cancer Hospital and Solove Research Institute at The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Haider Mahdi
- Division of Gynecologic Oncology, Obstetrics, Gynecology and Women's Health Institute, Cleveland Clinic, Cleveland, OH, United States.,Translational Hematology Oncology Research Department, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, United States.,Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| |
Collapse
|
113
|
Zhao Y, Liu L, Weng L. Comparisons of Underlying Mechanisms, Clinical Efficacy and Safety Between Anti-PD-1 and Anti-PD-L1 Immunotherapy: The State-of-the-Art Review and Future Perspectives. Front Pharmacol 2021; 12:714483. [PMID: 34305619 PMCID: PMC8293989 DOI: 10.3389/fphar.2021.714483] [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] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 06/24/2021] [Indexed: 12/13/2022] Open
Abstract
Over the past decade, diverse PD-1/PD-L1 blockades have demonstrated significant clinical benefit in across a wide range of tumor and cancer types. With the increasing number of PD-1/PD-L1 blockades available in the market, differences between the clinical performance of each of them started to be reported. Here, we provide a comprehensive historical and biological perspective regarding the underlying mechanism and clinical performance of PD-1/PD-L1 blockades, with an emphasis on the comparisons of their clinical efficacy and safety. The real-world evidence indicated that PD-1 blockade may be more effective than the PD-L1, though no significant differences were found as regards to their safety profiles. Future head-to-head studies are warranted for direct comparison between them. Finally, we summarize the yet to be elucidated questions and future promise of anti-PD-1/PD-L1 immunotherapy, including a need to explore novel biomarkers, novel combinatorial strategies, and their clinical use on chronic infection.
Collapse
Affiliation(s)
- Yating Zhao
- Institute of Pharmaceutical Science, King's College London, London, United Kingdom.,Clinical Pharmacology, BeiGene Ltd., Shanghai, China
| | - Liu Liu
- Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China
| | - Liang Weng
- Key Laboratory of Molecular Radiation Oncology, Changsha, China.,Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha, China.,Xiangya Lung Cancer Center, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
114
|
Dingemans AMC, Früh M, Ardizzoni A, Besse B, Faivre-Finn C, Hendriks LE, Lantuejoul S, Peters S, Reguart N, Rudin CM, De Ruysscher D, Van Schil PE, Vansteenkiste J, Reck M, on behalf of the ESMO Guidelines Committee. Small-cell lung cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up ☆. Ann Oncol 2021; 32:839-853. [PMID: 33864941 PMCID: PMC9464246 DOI: 10.1016/j.annonc.2021.03.207] [Citation(s) in RCA: 335] [Impact Index Per Article: 83.8] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 03/23/2021] [Accepted: 03/30/2021] [Indexed: 12/17/2022] Open
Affiliation(s)
- A.-M. C. Dingemans
- Department of Pulmonology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Department of Respiratory Medicine, Rotterdam
- Department of Pulmonology, GROW School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - M. Früh
- Department of Oncology and Haematology, Kantonsspital St. Gallen, St. Gallen
- Department of Medical Oncology, University of Bern, Bern, Switzerland
| | - A. Ardizzoni
- Division of Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - B. Besse
- Gustave Roussy, Villejuif
- Paris-Saclay University, Orsay, France
| | - C. Faivre-Finn
- Division of Cancer Sciences, University of Manchester & The Christie, NHS Foundation Trust, Manchester, UK
| | - L. E. Hendriks
- Department of Pulmonology, GROW School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - S. Lantuejoul
- Department of Biopathology, Centre Léon Bérard, Grenoble Alpes University, Lyon, France
| | - S. Peters
- Department of Oncology, Centre Hospitalier Universitaire Vaudois, Lausanne University, Lausanne, Switzerland
| | - N. Reguart
- Department of Medical Oncology, Hospital Clínic and Translational Genomics and Targeted Therapeutics in Solid Tumors, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - C. M. Rudin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, USA
| | - D. De Ruysscher
- Department of Radiation Oncology (Maastro Clinic), GROW School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - P. E. Van Schil
- Department of Thoracic and Vascular Surgery, Antwerp University Hospital and University of Antwerp, Edegem, Belgium
| | - J. Vansteenkiste
- Department of Respiratory Oncology, University Hospital KU Leuven, Leuven, Belgium
| | - M. Reck
- Department of Thoracic Oncology, Airway Research Center North, German Center for Lung Research, Lung Clinic, Grosshansdorf, Germany
| | | |
Collapse
|
115
|
Fu J, Su X, Li Z, Deng L, Liu X, Feng X, Peng J. HGF/c-MET pathway in cancer: from molecular characterization to clinical evidence. Oncogene 2021; 40:4625-4651. [PMID: 34145400 DOI: 10.1038/s41388-021-01863-w] [Citation(s) in RCA: 116] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 05/17/2021] [Accepted: 05/24/2021] [Indexed: 02/07/2023]
Abstract
This review provides a comprehensive landscape of HGF/c-MET (hepatocyte growth factor (HGF) /mesenchymal-epithelial transition factor (c-MET)) signaling pathway in cancers. First, we generalize the compelling influence of HGF/c-MET pathway on multiple cellular processes. Then, we present the genomic characterization of HGF/c-MET pathway in carcinogenesis. Furthermore, we extensively illustrate the malignant biological behaviors of HGF/c-MET pathway in cancers, in which hyperactive HGF/c-MET signaling is considered as a hallmark. In addition, we investigate the current clinical trials of HGF/c-MET-targeted therapy in cancers. We find that although HGF/c-MET-targeted therapy has led to breakthroughs in certain cancers, monotherapy of targeting HGF/c-MET has failed to demonstrate significant clinical efficacy in most cancers. With the advantage of the combinations of HGF/c-MET-targeted therapy, the exploration of more options of combinational targeted therapy in cancers may be the major challenge in the future.
Collapse
Affiliation(s)
- Jianjiang Fu
- Department of Pathology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- The Third Clinical School of Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Guangzhou, China
| | - Xiaorui Su
- Department of Pathology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- The Third Clinical School of Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Guangzhou, China
| | - Zhihua Li
- The Third Clinical School of Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Guangzhou, China
- Department of Fetal Medicine and Prenatal Diagnosis, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ling Deng
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiawei Liu
- Department of Pathology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- The Third Clinical School of Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Guangzhou, China
| | - Xuancheng Feng
- Department of Pathology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
- The Third Clinical School of Guangzhou Medical University, Guangzhou, China.
- Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Guangzhou, China.
| | - Juan Peng
- Department of Pathology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
- The Third Clinical School of Guangzhou Medical University, Guangzhou, China.
- Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Guangzhou, China.
| |
Collapse
|
116
|
Notch signaling and efficacy of PD-1/PD-L1 blockade in relapsed small cell lung cancer. Nat Commun 2021; 12:3880. [PMID: 34162872 PMCID: PMC8222224 DOI: 10.1038/s41467-021-24164-y] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 06/01/2021] [Indexed: 12/26/2022] Open
Abstract
Immune checkpoint blockade (ICB) benefits only a small subset of patients with small cell lung cancer (SCLC), yet the mechanisms driving benefit are poorly understood. To identify predictors of clinical benefit to ICB, we performed immunogenomic profiling of tumor samples from patients with relapsed SCLC. Tumors of patients who derive clinical benefit from ICB exhibit cytotoxic T-cell infiltration, high expression of antigen processing and presentation machinery (APM) genes, and low neuroendocrine (NE) differentiation. However, elevated Notch signaling, which positively correlates with low NE differentiation, most significantly predicts clinical benefit to ICB. Activation of Notch signaling in a NE human SCLC cell line induces a low NE phenotype, marked by increased expression of APM genes, demonstrating a mechanistic link between Notch activation, low NE differentiation and increased intrinsic tumor immunity. Our findings suggest Notch signaling as a determinant of response to ICB in SCLC. Immune checkpoint blockade (ICB) benefits only a small subset of patients with small cell lung cancer (SCLC) and the mechanisms driving benefit are poorly understood. Here, the authors show that elevated Notch signaling predicts clinical benefit in ICB in relapsed SCLC.
Collapse
|
117
|
Immunotherapy era in the treatment of small cell lung cancer. Med Oncol 2021; 38:86. [PMID: 34156575 DOI: 10.1007/s12032-021-01535-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 06/08/2021] [Indexed: 10/21/2022]
Abstract
Small cell lung cancer (SCLC) is differentiated from non-small cell lung cancers with its histological and morphological features, rapid response to chemotherapy, and recurrence in a short time after treatment is discontinued. Platinum plus etoposide chemotherapy combination has been used as a standard treatment, and no new drug has been found for more than 30 years in this disease. In research, the targeted pathways that may affect survival have not been identified yet. During the second half of the second decade of the 2000s, with immunotherapies that inhibit immune checkpoints, improvements in survival were achieved for the first time in treating SCLC after many years. Then a rapid increase was observed in chemotherapy plus immunotherapy combination studies in this field. Updated analyses of these studies were represented at international oncology meetings in 2020. Here, we reviewed immunotherapy studies conducted in patients with SCLC and the reflections on the daily practice.
Collapse
|
118
|
Abstract
OPINION STATEMENT Treatment recommendations for advanced gastroenteropancreatic neuroendocrine carcinomas (GEP-NEC) are based on uncontrolled, mainly retrospective data. Chemotherapy can offer palliative relief, but long-lasting complete responses or cures are rare. The European Neuroendocrine Tumour Society (ENETS) and European Society for Medical Oncology (ESMO) recommend platinum-based chemotherapy as first-line treatment. This has been the golden standard since the late 1980s and has been evaluated in mostly retrospective clinical studies. However, progression is inevitable for most patients. Unfortunately, data on effective second-line treatment options are scant, and ENETS and ESMO recommendations propose fluorouracil- or temozolomide-based chemotherapy schedules. As such, there is a huge unmet need for improved care. Improved knowledge on GEP-NEC biology may provide a pathway towards more effective interventions including chemotherapy, targeted gene therapy, peptide receptor radionuclide therapy, as well as immune checkpoint inhibitors. The review summarises this current state of the art as well as the most promising developments for systemic therapy in GEP-NEC patients.
Collapse
|
119
|
Brahmer JR, Abu-Sbeih H, Ascierto PA, Brufsky J, Cappelli LC, Cortazar FB, Gerber DE, Hamad L, Hansen E, Johnson DB, Lacouture ME, Masters GA, Naidoo J, Nanni M, Perales MA, Puzanov I, Santomasso BD, Shanbhag SP, Sharma R, Skondra D, Sosman JA, Turner M, Ernstoff MS. Society for Immunotherapy of Cancer (SITC) clinical practice guideline on immune checkpoint inhibitor-related adverse events. J Immunother Cancer 2021; 9:e002435. [PMID: 34172516 PMCID: PMC8237720 DOI: 10.1136/jitc-2021-002435] [Citation(s) in RCA: 446] [Impact Index Per Article: 111.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/24/2021] [Indexed: 02/06/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) are the standard of care for the treatment of several cancers. While these immunotherapies have improved patient outcomes in many clinical settings, they bring accompanying risks of toxicity, specifically immune-related adverse events (irAEs). There is a need for clear, effective guidelines for the management of irAEs during ICI treatment, motivating the Society for Immunotherapy of Cancer (SITC) to convene an expert panel to develop a clinical practice guideline. The panel discussed the recognition and management of single and combination ICI irAEs and ultimately developed evidence- and consensus-based recommendations to assist medical professionals in clinical decision-making and to improve outcomes for patients.
Collapse
Affiliation(s)
- Julie R Brahmer
- Department of Oncology and the Thoracic Oncology, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, Maryland, USA
| | - Hamzah Abu-Sbeih
- Department of Internal Medicine, University of Missouri-Kansas City, Kansas City, Missouri, USA
| | - Paolo Antonio Ascierto
- Unit of Melanoma Cancer Immunotherapy and Innovative Therapy, National Tumour Institute IRCCS Fondazione 'G. Pascale', Napoli, Italy
| | - Jill Brufsky
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Laura C Cappelli
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Frank B Cortazar
- Massachusetts General Hospital, Boston, Massachusetts, USA
- New York Nephrology Vasculitis and Glomerular Center, Albany, New York, USA
| | - David E Gerber
- Department of Hematology and Oncology, Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Lamya Hamad
- Department of Pharmacy, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Eric Hansen
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Douglas B Johnson
- Department of Medicine, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee, USA
| | - Mario E Lacouture
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Gregory A Masters
- Department of Medicine, Helen F. Graham Cancer Center, Newark, Delaware, USA
| | - Jarushka Naidoo
- Department of Oncology and the Thoracic Oncology, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, Maryland, USA
- Department of Oncology, Beaumont Hospital Dublin, The Royal College of Surgeons of Ireland, Dublin, Ireland
| | - Michele Nanni
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Miguel-Angel Perales
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Igor Puzanov
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Bianca D Santomasso
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Satish P Shanbhag
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Cancer Specialist of North Florida, Fleming Island, Florida, USA
| | - Rajeev Sharma
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Dimitra Skondra
- Department of Ophthalmology and Visual Science, University of Chicago Medical Center, Chicago, Illinois, USA
| | - Jeffrey A Sosman
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Medical Center, Chicago, Illinois, USA
| | - Michelle Turner
- Department of Oncology and the Thoracic Oncology, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore, Maryland, USA
| | - Marc S Ernstoff
- Division of Cancer Treatment & Diagnosis, National Cancer Institute, Rockville, Maryland, USA
| |
Collapse
|
120
|
Dumoulin DW, Dingemans AMC, Aerts JGJV, Remon J, De Ruysscher DKM, Hendriks LEL. Immunotherapy in small cell lung cancer: one step at a time: a narrative review. Transl Lung Cancer Res 2021; 10:2970-2987. [PMID: 34295691 PMCID: PMC8264327 DOI: 10.21037/tlcr-20-630] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 10/21/2020] [Indexed: 12/14/2022]
Abstract
Chemotherapy with or without radiotherapy has been the standard of care for many years for patients with small cell lung cancer (SCLC). Despite exceptionally high responses (up to 80%) with chemotherapy, the majority of patients relapse rapidly within weeks to months after treatment completion. Therefore, new and better treatment options are necessary. Recently, synergistic activity has been reported for the addition of immune checkpoint inhibitors (ICI) to standard platinum-based chemotherapy in the therapeutic strategy of advanced SCLC. For the first time after several decades, a significant survival improvement was achieved for this population. However, the overwhelming majority of patients do not respond to ICI, or relapse rapidly. There is need for better knowledge about the biology, histopathologic features, and molecular pathways of SCLC. This can probably help to identify the optimal predictive biomarkers, which are warranted to develop an individual therapeutic strategy including the rational use of a combination of immunotherapeutic agents. Here, we provide an overview of the rationale for and clinical results of the completed and ongoing trials using different strategies of immunotherapy in SCLC. In addition, opportunities for further improvement of therapies will be discussed, including the addition of radiotherapy, co-stimulatory antibodies, and other immune modifying agents.
Collapse
Affiliation(s)
- Daphne W. Dumoulin
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Anne-Marie C. Dingemans
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
- Department of Respiratory Medicine, Maastricht University Medical Centre, GROW School for Oncology and Developmental Biology, Maastricht, The Netherlands
| | - Joachim G. J. V. Aerts
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Jordi Remon
- Department of Medical Oncology, Centro Integral Oncológico Clara Campal Barcelona (CIOCCB), Hospital HM Delfos, HM Hospitales, Barcelona, Spain
| | - Dirk K. M. De Ruysscher
- Department of Radiation Oncology (MAASTRO Clinic), Maastricht University Medical Centre, GROW School for Oncology and Developmental Biology, Maastricht, The Netherlands
| | - Lizza E. L. Hendriks
- Department of Respiratory Medicine, Maastricht University Medical Centre, GROW School for Oncology and Developmental Biology, Maastricht, The Netherlands
| |
Collapse
|
121
|
Calvo E, Spira A, Miguel MD, Kondo S, Gazzah A, Millward M, Prenen H, Rottey S, Warburton L, Alanko T, Cassier PA, Yoh K, Italiano A, Moreno V, Peltola K, Seto T, Toyozawa R, Afar DE, Englert S, Komarnitsky P, Lambert S, Parikh A, Vosganian G, Gao B. Safety, pharmacokinetics, and efficacy of budigalimab with rovalpituzumab tesirine in patients with small cell lung cancer. Cancer Treat Res Commun 2021; 28:100405. [PMID: 34329846 DOI: 10.1016/j.ctarc.2021.100405] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/17/2021] [Accepted: 05/20/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Agents targeting programmed cell death protein 1 (PD-1) have been approved as monotherapy for patients with small cell lung cancer (SCLC). In preclinical models, the combined targeting of PD-1 and delta-like protein 3 resulted in enhanced antitumor activity. Herein, we report results from the expansion arm of study NCT03000257 evaluating the combination of the anti-PD-1 antibody budigalimab and the targeted antibody-drug conjugate rovalpituzumab tesirine (Rova-T) in patients with previously treated SCLC. MATERIALS AND METHODS This expansion arm of a multicenter, open-label, multi-arm, first-in-human phase 1 clinical trial enrolled adult patients with progressive SCLC. The primary objective was to assess safety and tolerability. Patients received budigalimab 375 mg via intravenous infusion every 3 weeks, and Rova-T was administered as a dose of 0.3 mg/kg intravenously, on day 1 of the first and third 3-week cycle. RESULTS As of October 2019, 31 patients with SCLC were enrolled and treated with budigalimab plus Rova-T. The combination was tolerated, with the most common treatment-emergent adverse events (in >30%) being pleural effusion, fatigue, and cough. The overall response rate was 24.1%, with one confirmed complete response and six confirmed partial responses. The overall response rate in patients with high delta-like protein 3 expression was similar (21.1%). The median progression-free survival was 3.48 months. CONCLUSION Combination therapy with budigalimab and Rova-T had promising efficacy and appeared to be tolerated in patients with SCLC. Although Rova-T development has been discontinued, development of budigalimab combined with other anticancer agents is ongoing. CLINICAL TRIAL REGISTRATION NUMBER NCT03000257 Statement on originality of the work The manuscript represents original work and has not been submitted for publication elsewhere nor previously published. Statement of prior presentation Data from this study were previously presented at the European Society for Medical Oncology (ESMO) Congress 2019.
Collapse
Affiliation(s)
- Emiliano Calvo
- START Madrid-CIOCC, Centro Integral Oncológico Clara Campal, Hospital Madrid Norte Sanchinarro, Calle Oña, 10, Madrid 28050, Spain.
| | - Alexander Spira
- Oncology Program, Virginia Cancer Specialists, Fairfax, VA, USA.
| | - María de Miguel
- START Madrid-CIOCC, Centro Integral Oncológico Clara Campal, Hospital Madrid Norte Sanchinarro, Calle Oña, 10, Madrid 28050, Spain.
| | - Shunsuke Kondo
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan.
| | - Anas Gazzah
- Department of Drug Development (DITEP), Gustave Roussy, Université Paris-Saclay, Villejuif, France.
| | | | - Hans Prenen
- Oncology Department, University Hospital Antwerp, Edegem, Belgium.
| | - Sylvie Rottey
- Drug Research Unit Ghent, Ghent University Hospital, Ghent, Belgium.
| | - Lydia Warburton
- Department of Medical Oncology, Sir Charles Gairdner Hospital, Nedlands, WA, Australia.
| | | | | | - Kiyotaka Yoh
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan.
| | - Antoine Italiano
- Early Phase Trials Unit, Institut Bergonié, University of Bordeaux, Bordeaux, France.
| | - Victor Moreno
- START Madrid-FJD, Hospital Universitario Fundación Jiménez Díaz, Madrid, Spain.
| | | | - Takashi Seto
- Department of Thoracic Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan
| | - Ryo Toyozawa
- Department of Thoracic Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan
| | | | | | | | | | | | | | - Bo Gao
- Cancer Care Center, Blacktown Hospital, Sydney, NSW, Australia.
| |
Collapse
|
122
|
Combination of the EP and Anti-PD-1 Pathway or Anti-CTLA-4 for the Phase III Trial of Small-Cell Lung Cancer: A Meta-Analysis. JOURNAL OF ONCOLOGY 2021; 2021:6662344. [PMID: 34122547 PMCID: PMC8166470 DOI: 10.1155/2021/6662344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 03/31/2021] [Accepted: 05/10/2021] [Indexed: 12/09/2022]
Abstract
The morbidity and mortality of lung cancer remain one of the highest among multiple cancers, respectively. Small-Cell Lung Cancer (SCLC) accounts for around 10%-15% of all lung cancers. Approximately two-thirds of the diagnosed SCLCs are in extensive stage (ES). Decades later, we still rely on the same traditional regimen with etoposide and platinum (EP) as the mainstay of treatment with poor prognosis. This meta-analysis aims to assess the effect of adding Immune Checkpoint Inhibitors (CPIs) such as (ipilimumab, atezolizumab, pembrolizumab, and durvalumab) to the traditional EP regimen for small-cell lung cancer extensive stage (ES-SCLC). We searched through PubMed looking for studies that compare between EP and CPIs, with EP alone, and only Phase III randomized controlled trials were considered eligible for this study. A total of 3645 papers were the results of the initial search, and only 4 studies met our criteria. Each investigator extracted the data independently using the PRISMA MODEL (Preferred Reporting Items for Systematic Reviews and Meta-Analyses) guideline. Each author used a prespecified sheet. The primary endpoint was to calculate OS (overall survival) and PFS (progression-free survival) hazard ratios for both arms. We found that adding EP plus CPIs increased both OS (HR, 95% CI 0.80 [0.70, 0.93], P = 0.0001, I 2 = 49%) and PFS (HR95% CI 0.81 [0.74, 0.88], P < 0.00001, I 2 = 0%). On the other hand, ORR (overall response rate) was not affected by the addition of CPIs to EP compared to EP alone, and the same was true for adverse events. To conclude, CTLA-4 alone is not encouraging, but PD-1/PD-L1 adds survival benefits. A combined treatment regimen shows to be more effective, improving overall survival rate Durvalumab and atezolizumab showed improvement for OS, but pembrolizumab and ipilimumab did not show a significant increase of OS over EP; however, pembrolizumab showed significant prolongation of the disease-free period.
Collapse
|
123
|
Shen Q, Qu J, Sheng L, Gao Q, Zhou J. Case Report: Transformation From Non-Small Cell Lung Cancer to Small Cell Lung Cancer During Anti-PD-1 Therapy: A Report of Two Cases. Front Oncol 2021; 11:619371. [PMID: 34094904 PMCID: PMC8176117 DOI: 10.3389/fonc.2021.619371] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 04/08/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Histological transformation of lung cancer to small cell lung cancer (SCLC) is uncommon. It is a small subset of the possible resistance mechanisms, even in epidermal growth factor receptor (EGFR)-mutated non-small cell lung cancer treated with EGFR-tyrosine kinase inhibitors. Reports on programmed cell death-1 (PD-1) inhibitors are rare. We report two cases of lung squamous carcinomas that transformed to SCLC during anti-PD-1 therapy, and present a detailed description of histological examination of the pre-and post-transformation tissues, hitherto absent from reports on the topic. CASE PRESENTATION Case 1: A 69-year-old man was diagnosed with stage IVa squamous cell carcinoma of the lung. He had a programmed cell death-ligand 1 tumor proportion score ≥50%. He achieved partial response after four cycles of sintilimab as first-line treatment. However, sintilimab was discontinued because of severe decrease in hemoglobin levels and platelet counts. Moreover, the occurrence of pleural effusion favored disease progression. Interestingly, bone marrow puncture and biopsy showed transformation to SCLC. Case 2: A 71-year-old man diagnosed with stage IIIa lung squamous cell carcinoma received neoadjuvant chemotherapy, underwent radical surgery, and finally received adjuvant chemotherapy. Five months later, he presented with tumor recurrence. He was treated with nivolumab, though disease progression was observed after four cycles. Notably, a subsequent computed tomography-guided biopsy showed SCLC. CONCLUSION Phenotypic transformation to SCLC is a potential mechanism of resistance to immunotherapy in squamous cell carcinomas of the lung. Disease progression should prompt re-biopsy to diagnose potential histological changes to assess the requirement for change in treatment.
Collapse
Affiliation(s)
- Qian Shen
- Department of Respiratory Disease, Thoracic Disease Centre, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jingjing Qu
- Department of Respiratory Disease, Thoracic Disease Centre, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Lingyan Sheng
- Department of Respiratory Disease, Thoracic Disease Centre, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Qiqi Gao
- Department of Pathology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jianying Zhou
- Department of Respiratory Disease, Thoracic Disease Centre, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
124
|
Biomarkers or factors for predicting the efficacy and adverse effects of immune checkpoint inhibitors in lung cancer: achievements and prospective. Chin Med J (Engl) 2021; 133:2466-2475. [PMID: 32960841 PMCID: PMC7575173 DOI: 10.1097/cm9.0000000000001090] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) are widely used in lung cancer therapy due to their effectiveness and minimal side effects. However, only a few lung cancer patients benefit from ICI therapy, driving the need to develop alternative biomarkers. Programmed death-ligand 1 (PD-L1) molecules expressed in tumor cells and immune cells play a key role in the immune checkpoint pathway. Therefore, PD-L1 expression is a prognostic biomarker in evaluating the effectiveness of programmed death-1 (PD-1)/PD-L1 inhibitors. Nevertheless, adverse predictive outcomes suggest that other factors are implicated in the response. In this review, we present a detailed introduction of existing biomarkers concerning tumor abnormality and host immunity. PD-L1 expression, tumor mutation burden, neoantigens, specific gene mutations, circulating tumor DNA, human leukocyte antigen class I, tumor microenvironment, peripheral inflammatory cells, and microbiome are discussed in detail. To sum up, this review provides information on the current application and future prospects of ICI biomarkers.
Collapse
|
125
|
|
126
|
Ouyang T, Cao Y, Kan X, Chen L, Ren Y, Sun T, Yan L, Xiong B, Liang B, Zheng C. Treatment-Related Serious Adverse Events of Immune Checkpoint Inhibitors in Clinical Trials: A Systematic Review. Front Oncol 2021; 11:621639. [PMID: 34046338 PMCID: PMC8144509 DOI: 10.3389/fonc.2021.621639] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 04/19/2021] [Indexed: 12/11/2022] Open
Abstract
Background Immune Checkpoint Inhibitors (ICI) have been progressively used in cancer treatment and produced unique toxicity profiles. This systematic review aims to comprehend the patterns and occurrence of treatment-related adverse events (trAEs) based on ICI. Methods PICOS/PRISMA methods were used to identify published English-language on PubMed, Web of Science, and Scopus from 2015 to 2020. Published clinical trials on ICI monotherapy, combined ICIs, and ICI plus other treatment with tabulated data on grade≥3 trAEs were included. Odds ratio (OR), χ2 tests were used to analyze for effect size and associations. Results This review included 145 clinical trials involving 21786 patients. Grade 3-5 trAEs were more common with ICI when they were plused with other treatments compared with ICI monotherapy(54.3% versus 17.7%, 46.1%, p<0.05). Grade 3-5 trAEs were also more common with CTLA-4 mAbs compared with anti-PD-1 and anti-PD-L1 (34.2% versus 15.1%, 13.6%, p<0.05). Hyperthyroidism (OR 3.8, 95%CI 1.7–8.6), nausea (OR 3.7, 95%CI 2.5–5.3), diarrhea (OR 2.7, 95%CI 2.2–3.2), colitis (OR 3.4, 95%CI 2.7–4.3), ALT increase (OR 4.9, 95%CI 3.9–6.1), AST increase (OR 3.8, 95%CI 3.0–4.9), pruritus (OR 2.4, 95%CI 1.5–3.9), rash (OR 2.8, 95%CI 2.1–3.8), fatigue (OR 2.8, 95%CI 2.2–3.7), decreased appetite (OR 2.4, 95%CI 1.5–3.8), and hypophysitis (OR 2.0, 95%CI 1.2–3.3) were more frequent with combined ICIs. Diarrhea (OR 8.1, 95%CI 6.4–10.3), colitis (OR 12.2, 95%CI 8.7–17.1), ALT increase (OR 5.1, 95%CI 3.5–7.4), AST increase (OR 4.2, 95%CI 2.8–6.3), pruritus (OR 4.1, 95%CI 2.0–8.4), rash (OR 4.4, 95%CI 2.9–6.8), hypophysitis (OR 12.1, 95%CI 6.3–23.4) were more common with CTLA-4 mAbs; whereas pneumonitis (OR 4.7, 95% CI 2.1–10.3) were more frequent with PD-1 mAbs. Conclusions Different immune checkpoint inhibitors are associated with different treatment-related adverse events profiles. A comprehensive data in this systematic review will provide comprehensive information for clinicians.
Collapse
Affiliation(s)
- Tao Ouyang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Yanyan Cao
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Xuefeng Kan
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Lei Chen
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Yanqiao Ren
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Tao Sun
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Liangliang Yan
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Bin Xiong
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Bin Liang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Chuansheng Zheng
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| |
Collapse
|
127
|
Stelwagen J, de Vries EGE, Walenkamp AME. Current Treatment Strategies and Future Directions for Extrapulmonary Neuroendocrine Carcinomas: A Review. JAMA Oncol 2021; 7:759-770. [PMID: 33630040 DOI: 10.1001/jamaoncol.2020.8072] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Importance Patients with extrapulmonary neuroendocrine carcinomas (EPNECs) receive essentially the same treatment as those with small cell lung cancer (SCLC) despite differences in origin, clinical course, and survival. This SCLC-based approach is attributable to the rarity of EPNECs, which impedes the use of randomized clinical trials. However, neuroendocrine carcinomas are becoming more common because of the increasing use of systemic cancer therapy for adenocarcinomas. This treatment can transdifferentiate certain adenocarcinomas into neuroendocrine carcinomas. In addition, the treatment landscape for SCLC is slowly changing, potentially impacting the treatment paradigms for EPNECs. Observations New information on tumorigenesis of EPNECs from different origins, either as a primary malignant tumor or after neuroendocrine differentiation from adenocarcinomas, demonstrates their biological similarity. Activated molecular pathways that appear to underlie the development of EPNECs are potentially targetable, and some of these targets, such as poly(adenosine diphosphate-ribose) polymerase, Wee1, and Aurora A kinase, are currently under investigation. Immune checkpoint inhibitors (ICIs) already constituted a new treatment modality for patients with SCLC and produced some promising results in patients with EPNECs. Conclusions and Relevance Although only moderately effective, the introduction of ICIs signifies the first new option in systemic treatment of SCLC in decades. To prove the value of ICIs and other new drugs for patients with EPNECs, these patients should be included in clinical trials independent of the primary tumor site. Furthermore, to optimize clinical decision-making for patients with EPNECs, experts from the neuroendocrine tumor board should collaborate with members from tumor site-specific boards, which will require patient referral to a center with EPNEC expertise.
Collapse
Affiliation(s)
- Johannes Stelwagen
- Department of Medical Oncology, University Medical Centre Groningen, University of Groningen, Groningen, the Netherlands
| | - Elisabeth G E de Vries
- Department of Medical Oncology, University Medical Centre Groningen, University of Groningen, Groningen, the Netherlands
| | - Annemiek M E Walenkamp
- Department of Medical Oncology, University Medical Centre Groningen, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
128
|
Chen S, Zhang Z, Zheng X, Tao H, Zhang S, Ma J, Liu Z, Wang J, Qian Y, Cui P, Huang D, Huang Z, Wu Z, Hu Y. Response Efficacy of PD-1 and PD-L1 Inhibitors in Clinical Trials: A Systematic Review and Meta-Analysis. Front Oncol 2021; 11:562315. [PMID: 33937012 PMCID: PMC8085334 DOI: 10.3389/fonc.2021.562315] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 03/19/2021] [Indexed: 12/13/2022] Open
Abstract
Background Immune checkpoint inhibitors targeting the PD-1/PD-L1 pathway have demonstrated promise in treating a variety of advanced cancers; however, little is known regarding their efficacy under various clinical situations, including different cancer types, treatment lines, drug combinations, and therapeutic regimens. Methods Published articles and conference abstracts (in English) in PubMed, Embase, the Cochrane Central Register, and Web of Science were searched up to February 10, 2020. The data were analyzed by the meta-analysis program in Stata. Results A total of 16,400 patients from 91 clinical trials were included in this meta-analysis. PD-1/PD-L1 inhibitors had a mean ORR of 19.56% (95% CI: 15.09–24.03), a median TTR of 2.05 months (m) (95%CI: 1.85–2.26), and a median DOR of 10.65 m (95%CI: 7.78–13.52). First-line treatment had a higher ORR (36.57% vs. 13.18%) but a shorter DOR (9.00 m vs. 13.42 m) compared to the second-line or subsequent treatment. Immunotherapy combined with chemotherapy (I+C) (46.81% [95%CI: 36.02–57.60]) had a statistically significant higher ORR compared to immunotherapy (I) (17.75% [95%CI: 14.47–21.03]) or immunotherapy combined with immunotherapy (I+O) (12.25% [95%CI: 1.56–22.94]), while I+C (8.09 m [95%CI: 6.86–9.32]) appeared to reduce the DOR compared to I (12.39 m [95%CI: 7.60–17.18]). PD-1 inhibitors were associated with better ORR (21.65% vs. 17.60%) and DOR (11.26 m vs. 10.03 m) compared to PD-L1 inhibitors. There were no significant differences in TTR under different situations. Conclusions PD-1/PD-L1 inhibitors were promising immunotherapeutic agents to achieve satisfactory response efficacies with different cancer types, treatment lines, drug combinations, and therapeutic regimens. This comprehensive summary of the response efficacy of PD-1/PD-L1 inhibitors serves as a reference for clinicians to make evidence-based decisions.
Collapse
Affiliation(s)
- Shixue Chen
- Department of Medical Oncology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China.,Department of Graduate Administration, Chinese PLA General Hospital, Beijing, China
| | - Zhibo Zhang
- Department of Cardiothoracic Surgery, The 78th Group Army Hospital of Chinese PLA, Mudanjiang, China
| | - Xuan Zheng
- Department of Medical Oncology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China.,Department of Graduate Administration, Chinese PLA General Hospital, Beijing, China
| | - Haitao Tao
- Department of Medical Oncology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Sujie Zhang
- Department of Medical Oncology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Junxun Ma
- Department of Medical Oncology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Zhefeng Liu
- Department of Medical Oncology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Jinliang Wang
- Department of Medical Oncology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Yuanyu Qian
- Department of Medical Oncology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Pengfei Cui
- Department of Medical Oncology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China.,Department of Graduate Administration, Chinese PLA General Hospital, Beijing, China
| | - Di Huang
- Department of Medical Oncology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China.,School of Medicine, Nankai University, Tianjin, China
| | - Ziwei Huang
- Department of Medical Oncology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China.,School of Medicine, Nankai University, Tianjin, China
| | - Zhaozhen Wu
- Department of Medical Oncology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China.,School of Medicine, Nankai University, Tianjin, China
| | - Yi Hu
- Department of Medical Oncology, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| |
Collapse
|
129
|
Thomas PL, Groves SM, Zhang YK, Li J, Gonzalez-Ericsson P, Sivagnanam S, Betts CB, Chen HC, Liu Q, Lowe C, Chen H, Boyd KL, Kopparapu PR, Yan Y, Coussens LM, Quaranta V, Tyson DR, Iams W, Lovly CM. Beyond Programmed Death-Ligand 1: B7-H6 Emerges as a Potential Immunotherapy Target in SCLC. J Thorac Oncol 2021; 16:1211-1223. [PMID: 33839362 DOI: 10.1016/j.jtho.2021.03.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 03/04/2021] [Accepted: 03/08/2021] [Indexed: 12/15/2022]
Abstract
INTRODUCTION The programmed death-ligand 1 (PD-L1) immune checkpoint inhibitors, atezolizumab and durvalumab, have received regulatory approval for the first-line treatment of patients with extensive-stage SCLC. Nevertheless, when used in combination with platinum-based chemotherapy, these PD-L1 inhibitors only improve overall survival by 2 to 3 months. This may be due to the observation that less than 20% of SCLC tumors express PD-L1 at greater than 1%. Evaluating the composition and abundance of checkpoint molecules in SCLC may identify molecules beyond PD-L1 that are amenable to therapeutic targeting. METHODS We analyzed RNA-sequencing data from SCLC cell lines (n = 108) and primary tumor specimens (n = 81) for expression of 39 functionally validated inhibitory checkpoint ligands. Furthermore, we generated tissue microarrays containing SCLC cell lines and patient with SCLC specimens to confirm expression of these molecules by immunohistochemistry. We annotated patient outcomes data, including treatment response and overall survival. RESULTS The checkpoint protein B7-H6 (NCR3LG1) exhibited increased protein expression relative to PD-L1 in cell lines and tumors (p < 0.05). Higher B7-H6 protein expression correlated with longer progression-free survival (p = 0.0368) and increased total immune infiltrates (CD45+) in patients. Furthermore, increased B7-H6 gene expression in SCLC tumors correlated with a decreased activated natural killer cell gene signature, suggesting a complex interplay between B7-H6 expression and immune signature in SCLC. CONCLUSIONS We investigated 39 inhibitory checkpoint molecules in SCLC and found that B7-H6 is highly expressed and associated with progression-free survival. In addition, 26 of 39 immune checkpoint proteins in SCLC tumors were more abundantly expressed than PD-L1, indicating an urgent need to investigate additional checkpoint targets for therapy in addition to PD-L1.
Collapse
Affiliation(s)
- Portia L Thomas
- Department of Microbiology, Immunology & Physiology, School of Medicine, Meharry Medical College, Nashville, Tennessee; School of Graduate Studies & Research, Meharry Medical College, Nashville, Tennessee
| | - Sarah M Groves
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee
| | - Yun-Kai Zhang
- Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jia Li
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Paula Gonzalez-Ericsson
- Breast Cancer Research Program, Vanderbilt Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee
| | - Shamilene Sivagnanam
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, Oregon; Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
| | - Courtney B Betts
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, Oregon; Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
| | - Hua-Chang Chen
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Qi Liu
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Cindy Lowe
- Department of Pathology, Immunology and Microbiology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Heidi Chen
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Kelli L Boyd
- Department of Pathology, Immunology and Microbiology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Prasad R Kopparapu
- Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Yingjun Yan
- Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Lisa M Coussens
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, Oregon; Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
| | - Vito Quaranta
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee
| | - Darren R Tyson
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee
| | - Wade Iams
- Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Christine M Lovly
- School of Graduate Studies & Research, Meharry Medical College, Nashville, Tennessee; Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee.
| |
Collapse
|
130
|
Xia C, Xu X, Ding Y, Yu C, Qiao J, Liu P. Abnormal spindle-like microcephaly-associated protein enhances cell invasion through Wnt/β-catenin-dependent regulation of epithelial-mesenchymal transition in non-small cell lung cancer cells. J Thorac Dis 2021; 13:2460-2474. [PMID: 34012593 PMCID: PMC8107535 DOI: 10.21037/jtd-21-566] [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] [Indexed: 12/15/2022]
Abstract
Background Lung cancer is one of the most common cancer worldwide, invasion and metastasis are still the bottleneck in the clinical setting. More diagnostic markers and drug targets need to be clarified. Therefore, we screened abnormal spindle-like microcephaly-associated protein (ASPM) as our candidate gene, which is associated with the poor prognosis. The aim of the present study was to understand the roles of ASPM in cell invasion in non-small cell lung cancer (NSCLC). Methods Gene Expression Omnibus (GEO) datamining was used to identify ASPM. Transwell invasion assay, quantitative reverse transcription polymerase chain reaction (qRT-PCR), and Western blot analysis were performed to discover the molecular functions of ASPM. Overexpression and small interfering mediated knockdown techniques have been used to study the cell invasion hallmarks of cancer. Results ASPM stood out among all the candidate genes from GEO datamining. ASPM in lung cancer tissues has been associated with poor overall survival rate. The protein levels of ASPM has been validated using lung cancer patients’ tissues, which upregulation of ASPM expression has been found in lung cancer patients. Silencing of ASPM decreased the cell invasion reflected by epithelial-mesenchymal transition (EMT) biomarkers: downregulation of vimentin and upregulation of E-cadherin. Matrix metalloproteinase (MMP) 2/9 protein levels were also affected upon transient knockdown of ASPM. Furthermore, the suppression of ASPM markedly inhibited the Wnt/β-catenin signaling pathway in vitro. The ectopic expression of ASPM had the opposite effect. The inhibition of β-catenin in ASPM-overexpressing lung cancer cells reduced the expression of EMT markers. The inhibitory effects on the Wnt/β-catenin signaling pathway were attenuated in cancer cells when ASPM was silenced. These findings demonstrated that the silencing of ASPM strongly reduced cell invasion in lung cancer. Conclusions ASPM promoted NSCLC invasion through EMT and by affecting the MMP family of proteins. The Wnt/β-catenin signaling pathway played an indispensable role in the ASPM-mediated NSCLC EMT-invasion cascade.
Collapse
Affiliation(s)
- Chunwei Xia
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Department of Respiratory Medicine, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaofeng Xu
- Department of Respiratory Medicine, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Yiyan Ding
- Department of Respiratory Medicine, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Cunjun Yu
- Department of Respiratory Medicine, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Jianbing Qiao
- Department of Respiratory Medicine, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Ping Liu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
131
|
Drapkin BJ, Rudin CM. Advances in Small-Cell Lung Cancer (SCLC) Translational Research. Cold Spring Harb Perspect Med 2021; 11:cshperspect.a038240. [PMID: 32513672 DOI: 10.1101/cshperspect.a038240] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Over the past several years, we have witnessed a resurgence of interest in the biology and therapeutic vulnerabilities of small-cell lung cancer (SCLC). This has been driven in part through the development of a more extensive array of representative models of disease, including a diverse variety of genetically engineered mouse models and human tumor xenografts. Herein, we review recent progress in SCLC model development, and consider some of the particularly active avenues of translational research in SCLC, including interrogation of intratumoral heterogeneity, insights into the cell of origin and oncogenic drivers, mechanisms of chemoresistance, and new therapeutic opportunities including biomarker-directed targeted therapies and immunotherapies. Whereas SCLC remains a highly lethal disease, these new avenues of translational research, bringing together mechanism-based preclinical and clinical research, offer new hope for patients with SCLC.
Collapse
Affiliation(s)
- Benjamin J Drapkin
- University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Charles M Rudin
- Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| |
Collapse
|
132
|
Schwendenwein A, Megyesfalvi Z, Barany N, Valko Z, Bugyik E, Lang C, Ferencz B, Paku S, Lantos A, Fillinger J, Rezeli M, Marko-Varga G, Bogos K, Galffy G, Renyi-Vamos F, Hoda MA, Klepetko W, Hoetzenecker K, Laszlo V, Dome B. Molecular profiles of small cell lung cancer subtypes: therapeutic implications. Mol Ther Oncolytics 2021; 20:470-483. [PMID: 33718595 PMCID: PMC7917449 DOI: 10.1016/j.omto.2021.02.004] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Small cell lung cancer (SCLC; accounting for approximately 13%-15% of all lung cancers) is an exceptionally lethal malignancy characterized by rapid doubling time and high propensity to metastasize. In contrast to the increasingly personalized therapies in other types of lung cancer, SCLC is still regarded as a homogeneous disease and the prognosis of SCLC patients remains poor. Recently, however, substantial progress has been made in our understanding of SCLC biology. Advances in genomics and development of new preclinical models have facilitated insights into the intratumoral heterogeneity and specific genetic alterations of this disease. This worldwide resurgence of studies on SCLC has ultimately led to the development of novel subtype-specific classifications primarily based on the neuroendocrine features and distinct molecular profiles of SCLC. Importantly, these biologically distinct subtypes might define unique therapeutic vulnerabilities. Herein, we summarize the current knowledge on the molecular profiles of SCLC subtypes with a focus on their potential clinical implications.
Collapse
Affiliation(s)
- Anna Schwendenwein
- Department of Thoracic Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, 1090 Vienna, Austria
| | - Zsolt Megyesfalvi
- Department of Thoracic Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, 1090 Vienna, Austria
- Department of Thoracic Surgery, National Institute of Oncology-Semmelweis University, 1122 Budapest, Hungary
- National Koranyi Institute of Pulmonology, 1121 Budapest, Hungary
| | - Nandor Barany
- Department of Thoracic Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, 1090 Vienna, Austria
- National Koranyi Institute of Pulmonology, 1121 Budapest, Hungary
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, 1085 Budapest, Hungary
| | - Zsuzsanna Valko
- Department of Thoracic Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, 1090 Vienna, Austria
- National Koranyi Institute of Pulmonology, 1121 Budapest, Hungary
| | - Edina Bugyik
- National Koranyi Institute of Pulmonology, 1121 Budapest, Hungary
| | - Christian Lang
- Department of Thoracic Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, 1090 Vienna, Austria
| | - Bence Ferencz
- Department of Thoracic Surgery, National Institute of Oncology-Semmelweis University, 1122 Budapest, Hungary
- National Koranyi Institute of Pulmonology, 1121 Budapest, Hungary
| | - Sandor Paku
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, 1085 Budapest, Hungary
| | - Andras Lantos
- National Koranyi Institute of Pulmonology, 1121 Budapest, Hungary
| | - Janos Fillinger
- Department of Thoracic Surgery, National Institute of Oncology-Semmelweis University, 1122 Budapest, Hungary
- National Koranyi Institute of Pulmonology, 1121 Budapest, Hungary
| | - Melinda Rezeli
- Department of Biomedical Engineering, Lund University, 221 00 Lund, Sweden
| | - Gyorgy Marko-Varga
- Department of Biomedical Engineering, Lund University, 221 00 Lund, Sweden
| | - Krisztina Bogos
- National Koranyi Institute of Pulmonology, 1121 Budapest, Hungary
| | - Gabriella Galffy
- Torokbalint County Institute of Pulmonology, 2045 Torokbalint, Hungary
| | - Ferenc Renyi-Vamos
- Department of Thoracic Surgery, National Institute of Oncology-Semmelweis University, 1122 Budapest, Hungary
- National Koranyi Institute of Pulmonology, 1121 Budapest, Hungary
| | - Mir Alireza Hoda
- Department of Thoracic Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, 1090 Vienna, Austria
| | - Walter Klepetko
- Department of Thoracic Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, 1090 Vienna, Austria
| | - Konrad Hoetzenecker
- Department of Thoracic Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, 1090 Vienna, Austria
| | - Viktoria Laszlo
- Department of Thoracic Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, 1090 Vienna, Austria
- Department of Thoracic Surgery, National Institute of Oncology-Semmelweis University, 1122 Budapest, Hungary
- National Koranyi Institute of Pulmonology, 1121 Budapest, Hungary
| | - Balazs Dome
- Department of Thoracic Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, 1090 Vienna, Austria
- Department of Thoracic Surgery, National Institute of Oncology-Semmelweis University, 1122 Budapest, Hungary
- National Koranyi Institute of Pulmonology, 1121 Budapest, Hungary
| |
Collapse
|
133
|
Baena J, Modrego A, Zeaiter A, Kahatt C, Alfaro V, Jimenez-Aguilar E, Mazarico JM, Paz-Ares L. Lurbinectedin in the treatment of relapsed small cell lung cancer. Future Oncol 2021; 17:2279-2289. [PMID: 33736462 DOI: 10.2217/fon-2020-1212] [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: 12/16/2022] Open
Abstract
Lurbinectedin is a marine-derived drug that inhibits transcription, a process that is frequently dysregulated in small cell lung cancer. The activity of lurbinectedin has been studied in many solid tumors, showing not only promising results but also a favorable safety profile. In relapsed small cell lung cancer, the drug has shown encouraging activity both as a single agent and in combination with doxorubicin, paclitaxel or irinotecan. The USA FDA has recently granted accelerated approval to lurbinectedin monotherapy in this setting. This article provides an update on available data and ongoing studies of lurbinectedin in small cell lung cancer, including Phase I combination trials, the basket Phase II trial and the ATLANTIS Phase III trial.
Collapse
Affiliation(s)
- Javier Baena
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, 28041, Spain.,H12O-CNIO Lung Cancer Clinical Research Unit, Instituto de Investigación Sanitaria Hospital Universitario 12 de Octubre, Spanish National Cancer Research Center, Madrid, 28029, Spain
| | - Andrea Modrego
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, 28041, Spain
| | - Ali Zeaiter
- Clinical Development, PharmaMar, Madrid, 28770, Spain
| | - Carmen Kahatt
- Clinical Development, PharmaMar, Madrid, 28770, Spain
| | | | - Elizabeth Jimenez-Aguilar
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, 28041, Spain.,H12O-CNIO Lung Cancer Clinical Research Unit, Instituto de Investigación Sanitaria Hospital Universitario 12 de Octubre, Spanish National Cancer Research Center, Madrid, 28029, Spain
| | - Jose María Mazarico
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, 28041, Spain.,H12O-CNIO Lung Cancer Clinical Research Unit, Instituto de Investigación Sanitaria Hospital Universitario 12 de Octubre, Spanish National Cancer Research Center, Madrid, 28029, Spain
| | - Luis Paz-Ares
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, 28041, Spain.,H12O-CNIO Lung Cancer Clinical Research Unit, Instituto de Investigación Sanitaria Hospital Universitario 12 de Octubre, Spanish National Cancer Research Center, Madrid, 28029, Spain.,Cancer Networking Biomedical Research Center, Madrid, 28029, Spain.,Faculty of Medicine, Complutense University, Madrid, 28040, Spain
| |
Collapse
|
134
|
Hou W, Zhou X, Yi C, Zhu H. Immune Check Point Inhibitors and Immune-Related Adverse Events in Small Cell Lung Cancer. Front Oncol 2021; 11:604227. [PMID: 33816235 PMCID: PMC8016392 DOI: 10.3389/fonc.2021.604227] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 02/15/2021] [Indexed: 02/05/2023] Open
Abstract
Small cell lung cancer (SCLC) is a malignant solid tumor. In recent years, although immune check point inhibitors (ICIs) have achieved important advances in the treatment of SCLC, immune-related adverse events (irAEs) have occurred at the same time during the therapeutic period. Some irAEs lead to dose reduction or treatment rejection. The immune microenvironment of SCLC is complicated, therefore, understanding irAEs associated with ICIs is of great importance and necessity for the clinical management of SCLC. However, the lack of comprehensive understanding of irAEs in patients with SCLC remains remarkable. This review aims to provide an up-to-date overview of ICIs and their associated irAEs in patients with SCLC based on present clinical data.
Collapse
Affiliation(s)
- Wanting Hou
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaohan Zhou
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Cheng Yi
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Hong Zhu
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
135
|
Neuroendocrine-Related Circulating Transcripts in Small-Cell Lung Cancers: Detection Methods and Future Perspectives. Cancers (Basel) 2021; 13:cancers13061339. [PMID: 33809582 PMCID: PMC8061767 DOI: 10.3390/cancers13061339] [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: 02/04/2021] [Revised: 02/28/2021] [Accepted: 03/11/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary The recent implementation of techniques to study circulating tumor cells allowed a rapid increase in knowledge about the molecular basis of Small-Cell Lung Cancer (SCLC), which appears to be more heterogeneous and dynamic than expected. Here, we present a summary of current knowledge and new findings about some of the neuroendocrine-related transcripts expressed in SCLC patients that could offer a great opportunity in distinguishing and managing different SCLC phenotypes. Abstract No well-established prognostic or predictive molecular markers of small-cell lung cancer (SCLC) are currently available; therefore, all patients receive standard treatment. Adequate quantities and quality of tissue samples are frequently unavailable to perform a molecular analysis of SCLC, which appears more heterogeneous and dynamic than expected. The implementation of techniques to study circulating tumor cells could offer a suitable alternative to expand the knowledge of the molecular basis of a tumor. In this context, the advantage of SCLC circulating cells to express some specific markers to be explored in blood as circulating transcripts could offer a great opportunity in distinguishing and managing different SCLC phenotypes. Here, we present a summary of published data and new findings about the detection methods and potential application of a group of neuroendocrine related transcripts in the peripheral blood of SCLC patients. In the era of new treatments, easy and rapid detection of informative biomarkers in blood warrants further investigation, since it represents an important option to obtain essential information for disease monitoring and/or better treatment choices.
Collapse
|
136
|
Owonikoko TK, Park K, Govindan R, Ready N, Reck M, Peters S, Dakhil SR, Navarro A, Rodríguez-Cid J, Schenker M, Lee JS, Gutierrez V, Percent I, Morgensztern D, Barrios CH, Greillier L, Baka S, Patel M, Lin WH, Selvaggi G, Baudelet C, Baden J, Pandya D, Doshi P, Kim HR. Nivolumab and Ipilimumab as Maintenance Therapy in Extensive-Disease Small-Cell Lung Cancer: CheckMate 451. J Clin Oncol 2021; 39:1349-1359. [PMID: 33683919 PMCID: PMC8078251 DOI: 10.1200/jco.20.02212] [Citation(s) in RCA: 190] [Impact Index Per Article: 47.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
In extensive-disease small-cell lung cancer (ED-SCLC), response rates to first-line platinum-based chemotherapy are robust, but responses lack durability. CheckMate 451, a double-blind phase III trial, evaluated nivolumab plus ipilimumab and nivolumab monotherapy as maintenance therapy following first-line chemotherapy for ED-SCLC.
Collapse
Affiliation(s)
| | - Keunchil Park
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Ramaswamy Govindan
- Alvin J Siteman Cancer Center at Washington University School of Medicine, St Louis, MO
| | - Neal Ready
- Duke University Medical Center, Durham, NC
| | - Martin Reck
- Department of Thoracic Oncology, Airway Research Center North, German Center for Lung Research, LungClinic, Grosshansdorf, Germany
| | - Solange Peters
- Oncology Department, Lausanne University Hospital, Lausanne, Switzerland
| | | | - Alejandro Navarro
- Vall d'Hebron University Hospital and Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Jerónimo Rodríguez-Cid
- Centro Oncológico, Médica Sur-Instituto Nacional de Enfermedades Respiratorias, Mexico City, Mexico
| | | | - Jong-Seok Lee
- Seoul National University Bundang Hospital, Seongnam, South Korea
| | | | | | - Daniel Morgensztern
- Alvin J Siteman Cancer Center at Washington University School of Medicine, St Louis, MO
| | - Carlos H Barrios
- Oncology Research Center, Hospital São Lucas, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Laurent Greillier
- Aix Marseille Univ, APHM, INSERM, CNRS, CRCM, Hôpital Nord, Multidisciplinary Oncology and Therapeutic Innovations Department, Marseille, France
| | - Sofia Baka
- Interbalkan European Medical Center, Thessaloniki, Greece
| | - Miten Patel
- Cancer Specialists of North Florida, Jacksonville, FL
| | | | | | | | | | | | | | - Hye Ryun Kim
- Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
137
|
Twomey JD, Zhang B. Cancer Immunotherapy Update: FDA-Approved Checkpoint Inhibitors and Companion Diagnostics. AAPS J 2021; 23:39. [PMID: 33677681 PMCID: PMC7937597 DOI: 10.1208/s12248-021-00574-0] [Citation(s) in RCA: 437] [Impact Index Per Article: 109.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 02/15/2021] [Indexed: 12/13/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) are considered a new standard-of-care across many cancer indications. This review provides an update on ICIs approved by the Food and Drug Administration (FDA), with focus on monoclonal antibodies that target the programmed cell death 1 (PD-1) or its ligand, PD-1 ligand 1 (PD-L1), including information on their clinical indications and associated companion diagnostics. The information is further discussed with strategies for identifying predictive biomarkers to guide the clinical use of PD-1/PD-L1-targeted therapies.
Collapse
Affiliation(s)
- Julianne D Twomey
- Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, 20993, USA.
| | - Baolin Zhang
- Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, 20993, USA.
| |
Collapse
|
138
|
Montenegro GB, Farid S, Liu SV. Immunotherapy in lung cancer. J Surg Oncol 2021; 123:718-729. [PMID: 33595888 DOI: 10.1002/jso.26347] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 11/20/2020] [Accepted: 11/21/2020] [Indexed: 12/22/2022]
Abstract
Immunotherapy has emerged as an important treatment modality throughout oncology with a particularly important role in the treatment of lung cancer. Early signals showed responses could be achieved in nonsmall cell lung cancer and small cell lung cancer and these monoclonal antibodies have become standards of care for advanced stage disease. They have also shown promise in earlier-stage disease as complements to radiation or surgery, offering the potential for durable, meaningful survival gains.
Collapse
Affiliation(s)
| | - Saira Farid
- Department of Medicine, Washington Hospital Center, Washington, District of Columbia, USA
| | - Stephen V Liu
- Division of Medical Oncology, Georgetown University, Washington, District of Columbia, USA
| |
Collapse
|
139
|
Giffin MJ, Cooke K, Lobenhofer EK, Estrada J, Zhan J, Deegen P, Thomas M, Murawsky CM, Werner J, Liu S, Lee F, Homann O, Friedrich M, Pearson JT, Raum T, Yang Y, Caenepeel S, Stevens J, Beltran PJ, Canon J, Coxon A, Bailis JM, Hughes PE. AMG 757, a Half-Life Extended, DLL3-Targeted Bispecific T-Cell Engager, Shows High Potency and Sensitivity in Preclinical Models of Small-Cell Lung Cancer. Clin Cancer Res 2021; 27:1526-1537. [PMID: 33203642 DOI: 10.1158/1078-0432.ccr-20-2845] [Citation(s) in RCA: 127] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 10/21/2020] [Accepted: 11/13/2020] [Indexed: 12/12/2022]
Abstract
PURPOSE Small-cell lung cancer (SCLC) is an aggressive neuroendocrine tumor with a high relapse rate, limited therapeutic options, and poor prognosis. We investigated the antitumor activity of AMG 757, a half-life extended bispecific T-cell engager molecule targeting delta-like ligand 3 (DLL3)-a target that is selectively expressed in SCLC tumors, but with minimal normal tissue expression. EXPERIMENTAL DESIGN AMG 757 efficacy was evaluated in SCLC cell lines and in orthotopic and patient-derived xenograft (PDX) mouse SCLC models. Following AMG 757 administration, changes in tumor volume, pharmacodynamic changes in tumor-infiltrating T cells (TILs), and the spatial relationship between the appearance of TILs and tumor histology were examined. Tolerability was assessed in nonhuman primates (NHPs). RESULTS AMG 757 showed potent and specific killing of even those SCLC cell lines with very low DLL3 expression (<1,000 molecules per cell). AMG 757 effectively engaged systemically administered human T cells, induced T-cell activation, and redirected T cells to lyse tumor cells to promote significant tumor regression and complete responses in PDX models of SCLC and in orthotopic models of established primary lung SCLC and metastatic liver lesions. AMG 757 was well tolerated with no AMG 757-related adverse findings up to the highest tested dose (4.5 mg/kg weekly) in NHP. AMG 757 exhibits an extended half-life in NHP, which is projected to enable intermittent administration in patients. CONCLUSIONS AMG 757 has a compelling safety and efficacy profile in preclinical studies making it a viable option for targeting DLL3-expressing SCLC tumors in the clinical setting.
Collapse
Affiliation(s)
| | - Keegan Cooke
- Oncology Research, Amgen Research, Thousand Oaks, California
| | - Edward K Lobenhofer
- Translational Safety & Bioanalytical Sciences, Amgen Research, Thousand Oaks, California
| | - Juan Estrada
- Oncology Research, Amgen Research, Thousand Oaks, California
| | - Jinghui Zhan
- Oncology Research, Amgen Research, Thousand Oaks, California
| | - Petra Deegen
- Translational Safety & Bioanalytical Sciences, Amgen Research (Munich) GmbH, Munich, Germany
| | - Melissa Thomas
- Therapeutic Discovery, Amgen Research, South San Francisco, California
| | | | - Jonathan Werner
- Translational Safety & Bioanalytical Sciences, Amgen Research, Thousand Oaks, California
| | - Siyuan Liu
- Oncology Research, Amgen Research, Thousand Oaks, California
| | - Fei Lee
- Oncology Research, Amgen Research, South San Francisco, California
| | - Oliver Homann
- Genome Analysis Unit, Amgen Research, South San Francisco, California
| | - Matthias Friedrich
- Translational Safety & Bioanalytical Sciences, Amgen Research (Munich) GmbH, Munich, Germany
| | - Joshua T Pearson
- Pharmacokinetics & Drug Metabolism, Amgen Research, South San Francisco, California
| | - Tobias Raum
- Therapeutic Discovery, Amgen Research (Munich) GmbH, Munich, Germany
| | - Yajing Yang
- Oncology Research, Amgen Research, Thousand Oaks, California
| | - Sean Caenepeel
- Oncology Research, Amgen Research, Thousand Oaks, California
| | - Jennitte Stevens
- Therapeutic Discovery, Amgen Research, Thousand Oaks, California
| | - Pedro J Beltran
- Oncology Research, Amgen Research, Thousand Oaks, California
| | - Jude Canon
- Oncology Research, Amgen Research, Thousand Oaks, California
| | - Angela Coxon
- Oncology Research, Amgen Research, Thousand Oaks, California
| | - Julie M Bailis
- Oncology Research, Amgen Research, South San Francisco, California.
| | - Paul E Hughes
- Oncology Research, Amgen Research, Thousand Oaks, California.
| |
Collapse
|
140
|
SchnÖller L, KÄsmann L, Taugner J, Abdo R, Eze C, Manapov F. Prognostic Role of Lung Immune Scores for Prediction of Survival in Limited-stage Small Cell Lung Cancer. In Vivo 2021; 35:929-935. [PMID: 33622885 PMCID: PMC8045058 DOI: 10.21873/invivo.12333] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 12/10/2020] [Accepted: 12/21/2020] [Indexed: 12/31/2022]
Abstract
BACKGROUND/AIM Previous studies have suggested the prognostic value of the Lung Immune Prediction Index (LIPI) and the Gustave Roussy Score (GRIM) as prognostic markers in advanced small cell lung cancer (SCLC). However, LIPI and GRIM score have not been evaluated in patients with limited stage SCLC (LS-SCLC). PATIENTS AND METHODS Pretreatment LIPI and GRIM score of 33 (43%) patients out of 77 LS-SCLC patients treated with chemoradiotherapy (CRT) during 2004-2015 were included. RESULTS The median overall survival (OS) time in the good, intermediate, and poor LIPI subgroups were 14, 17 and 3 months (p=0.973) and 14, 17 and 17 months in the GRIM subgroups. In univariate analysis, patients age <65 years (p=0.008), concurrent chemotherapy (p=0.028), and administering prophylactic cranial irradiation (PCI) (p=0.031) were associated with improved OS. Using Cox regression analysis, age remained significant (HR=3.299, p=0.031) and PCI showed a trend (HR=2.801, p=0.06). CONCLUSION Independent predictors of overall survival were identified and can contribute to improved treatment personalization. Concurrent chemotherapy and PCI after CRT were associated with improved OS compared to LIPI- and GRIM-score, which had no prognostic impact in LS-SCLC.
Collapse
Affiliation(s)
- Leon SchnÖller
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Lukas KÄsmann
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany;
- Comprehensive Pneumology Center Munich (CPC-M), Member of the German Center for Lung Research (DZL), Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Julian Taugner
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Reem Abdo
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Chukwuka Eze
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Farkhad Manapov
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
- Comprehensive Pneumology Center Munich (CPC-M), Member of the German Center for Lung Research (DZL), Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| |
Collapse
|
141
|
Zhang B, Birer SR, Dvorkin M, Shruti J, Byers L. New Therapies and Biomarkers: Are We Ready for Personalized Treatment in Small Cell Lung Cancer? Am Soc Clin Oncol Educ Book 2021; 41:1-10. [PMID: 33979194 DOI: 10.1200/edbk_320673] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Small cell lung cancer (SCLC) is an aggressive form of lung cancer with a 5-year survival rate of less than 7%. In contrast to non-small cell lung cancer, SCLC has long been treated as a homogeneous disease without personalized treatment options. In recent years, the incorporation of immunotherapy into the treatment paradigm has brought moderate benefit to patients with SCLC; however, more effective therapies are urgently needed. In this article, we describe the current treatment standards and emerging therapeutic approaches for the treatment of SCLC. We also discuss promising biomarkers in SCLC and the recently discovered four subtypes of SCLC, each with its unique therapeutic vulnerability. Lastly, we discuss the advances in radiation therapy for the treatment of SCLC.
Collapse
Affiliation(s)
- Bingnan Zhang
- Division of Cancer Medicine, MD Anderson Cancer Center, Houston, TX
| | - Samuel R Birer
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI
| | - Mikhail Dvorkin
- BHI of Omsk Region Clinical Oncology Dispensary, Omsk, Russia
| | - Jolly Shruti
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI
| | - Lauren Byers
- Department of Thoracic/Head and Neck Medical Oncology, MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
142
|
History of Extensive Disease Small Cell Lung Cancer Treatment: Time to Raise the Bar? A Review of the Literature. Cancers (Basel) 2021; 13:cancers13050998. [PMID: 33673630 PMCID: PMC7957518 DOI: 10.3390/cancers13050998] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/11/2021] [Accepted: 02/19/2021] [Indexed: 12/31/2022] Open
Abstract
Simple Summary Small cell lung cancer (SCLC) remains the most aggressive form of neuroendocrine tumor of the lung, for which treatment options remain limited. The introduction of immune checkpoint inhibitors has modified for the first time the therapeutic strategies in patients with extensive disease after decades. New therapeutic approaches are required. Deeper knowledge of tumor biology is required to gain new insights into this complex disease. Abstract Several trials have tried for decades to improve the outcome of extensive disease small cell lung cancer (ED-SCLC) through attempts to modify the standard treatments. Nevertheless, platinum/etoposide combination and topotecan have remained respectively the first and the second line standard treatments for the last 40 years. With the advent of immunotherapy, this scenario has finally changed. Our review aims to provide an overview of the primary studies on the actual therapeutic strategies available for ED-SCLC patients, and to highlight emerging evidence supporting the use of immunotherapy in SCLC patients.
Collapse
|
143
|
Yang Y, Yang L, Wang Y. [Immunotherapy for Lung Cancer: Mechanisms of Resistance and Response Strategy]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2021; 24:112-123. [PMID: 33626853 PMCID: PMC7936078 DOI: 10.3779/j.issn.1009-3419.2021.101.02] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Inhibition of immune checkpoints is at the forefront of immunotherapy for lung cancer. However, a high percentage of lung cancer patients do not respond to these immunotherpy or their responses are transient, indicating the existence of immune resistance. Emerging evidence suggested that the interactions between cancer cells and immune system were continuous and dynamic. Here, we review how a range of cancer-cell-autonomous characteristics, tumor-microenvironment factors, and host-related influences account for heterogenous responses. Furthermore, with the identification of new targets of immunotherapy and development of immune-based combination therapy, we elucidate the methods might useful to overcome resistance.
Collapse
Affiliation(s)
- Yaning Yang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital,
Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Lu Yang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital,
Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yan Wang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital,
Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| |
Collapse
|
144
|
He Z, Zhou H, Wang J, Li D, Zhang X, Wang P, Ma T, Zhang Y, Tian C, Chen Y, Zou M, Han Y, Xu C, Ma S, Wang L, Wu X, Chen G, Wang Q. Apatinib with etoposide capsules as a third- or further-line therapy for extensive-stage small cell lung cancer: an open-label, multicenter, single-arm phase II trial. Transl Lung Cancer Res 2021; 10:889-899. [PMID: 33718030 PMCID: PMC7947412 DOI: 10.21037/tlcr-20-1235] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Background Patients with extensive-stage small-cell lung cancer (ES-SCLC) have a particularly poor prognosis. And the treatment options for patients with relapsed or refractory ES-SCLC are limited. Thus, we conducted an open-label, multicenter, single-arm phase II clinical trial to assess the efficacy and safety of apatinib plus etoposide capsules as the third- or further-line treatment in ES-SCLC patients. Methods Patients with ES-SCLC who experienced disease progression following 2 to 3 previous therapies from 11 medical centers in China were enrolled to receive apatinib (250 mg/d, continuously) and etoposide capsules (50 mg/d, on day 1–21, per 28 days). The treatment continued until disease progression, treatment intolerance, or death. The primary endpoint was progression-free survival (PFS), and the secondary endpoints were objective response rate (ORR), overall survival (OS), and safety. Results Fifty-six patients with relapsed or refractory ES-SCLC were enrolled from January 2018 to February 2020 and 53 of them were eventually included in the evaluation population. The median follow-up was 9.8 months. At the data cut-off time (March 5, 2020), 39 patients (74%) had died and 44 (83%) had progressed. The median PFS was 3.0 months (95% CI, 2.1–3.9) and the median OS was 5.0 months (95% CI, 3.6–6.4). No complete responses were seen. Eleven patients (21%) showed a best response of partial response and 37 (70%) patients achieved stable disease. The ORR was 20.8% (11/53), and the disease control rate (DCR) was 90.6% (48/53). The 6-month OS rate was 40.1% (95% CI, 26.2–54). After 12 months, the OS rate was 18.4% (95% CI, 4.7–32.1). Possible treatment-related grade III/IV adverse events included leukopenia [8 (15.1%)], neutropenia [7 (13.2%)], anemia [4 (7.4%)], and hand-foot syndrome [2 (3.8%)]. During the study, no mortality occurred as a consequence of treatment. Conclusions Apatinib combined with etoposide capsules exhibits efficacy and has an acceptable safety profile. It could be used as a later-line treatment for ES-SCLC patients who have been heavily pretreated with standard therapies. Further exploration of apatinib combined with etoposide capsules in phase III trials is warranted.
Collapse
Affiliation(s)
- Zhen He
- Department of Internal Medicine, Henan Cancer Hospital, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Hanqiong Zhou
- Department of Internal Medicine, Henan Cancer Hospital, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Junsheng Wang
- Department of Medical Oncology, Anyang Cancer Hospital, Anyang, China
| | - Ding Li
- Department of Pharmacy, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Xudong Zhang
- Department of Medical Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Pengyuan Wang
- Department of Medical Oncology, Xuchang Central Hospital, Xuchang, China
| | - Tianjiang Ma
- Department of Medical Oncology, Luohe Central Hospital, Luohe, China
| | - Yueqiang Zhang
- Department of Medical Oncology, Zhoukou Chinese Medicine Hospital, Zhoukou, China
| | - Chuntao Tian
- Department of Medical Oncology, Sanmenxia Central Hospital, Sanmenxia, China
| | - Yunfang Chen
- Department of Medical Oncology, Zhumadian Central Hospital, Zhumadian, China
| | - Minglei Zou
- Department of Medical Oncology, The Second People's Hospital of Jiaozuo, Jiaozuo, China
| | - Yu Han
- Department of Medical Oncology, Jiyuan People's Hospital of Henan Province, Jiyuan, China
| | - Cong Xu
- Department of Internal Medicine, Henan Cancer Hospital, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Shuxiang Ma
- Department of Internal Medicine, Henan Cancer Hospital, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Lili Wang
- Department of Internal Medicine, Henan Cancer Hospital, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Xuan Wu
- Department of Internal Medicine, Henan Cancer Hospital, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Gongbin Chen
- Department of Medical Oncology, Shangqiu first People's Hospital, Shangqiu, China
| | - Qiming Wang
- Department of Internal Medicine, Henan Cancer Hospital, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
145
|
Zhou F, Qiao M, Zhou C. The cutting-edge progress of immune-checkpoint blockade in lung cancer. Cell Mol Immunol 2021; 18:279-293. [PMID: 33177696 PMCID: PMC8027847 DOI: 10.1038/s41423-020-00577-5] [Citation(s) in RCA: 135] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 10/15/2020] [Indexed: 12/24/2022] Open
Abstract
Great advances in immune checkpoint blockade have resulted in a paradigm shift in patients with lung cancer. Immune-checkpoint inhibitor (ICI) treatment, either as monotherapy or combination therapy, has been established as the standard of care for patients with locally advanced/metastatic non-small cell lung cancer without EGFR/ALK alterations or extensive-stage small cell lung cancer. An increasing number of clinical trials are also ongoing to further investigate the role of ICIs in patients with early-stage lung cancer as neoadjuvant or adjuvant therapy. Although PD-L1 expression and tumor mutational burden have been widely studied for patient selection, both of these biomarkers are imperfect. Due to the complex cancer-immune interactions among tumor cells, the tumor microenvironment and host immunity, collaborative efforts are needed to establish a multidimensional immunogram to integrate complementary predictive biomarkers for personalized immunotherapy. Furthermore, as a result of the wide use of ICIs, managing acquired resistance to ICI treatment remains an inevitable challenge. A deeper understanding of the underlying biological mechanisms of acquired resistance to ICIs is helpful to overcome these obstacles. In this review, we describe the cutting-edge progress made in patients with lung cancer, the optimal duration of ICI treatment, ICIs in some special populations, the unique response patterns during ICI treatment, the emerging predictive biomarkers, and our understanding of primary and acquired resistance mechanisms to ICI treatment.
Collapse
Affiliation(s)
- Fei Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Meng Qiao
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Caicun Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
146
|
Spigel DR, Vicente D, Ciuleanu TE, Gettinger S, Peters S, Horn L, Audigier-Valette C, Pardo Aranda N, Juan-Vidal O, Cheng Y, Zhang H, Shi M, Luft A, Wolf J, Antonia S, Nakagawa K, Fairchild J, Baudelet C, Pandya D, Doshi P, Chang H, Reck M. Second-line nivolumab in relapsed small-cell lung cancer: CheckMate 331 ☆. Ann Oncol 2021; 32:631-641. [PMID: 33539946 DOI: 10.1016/j.annonc.2021.01.071] [Citation(s) in RCA: 192] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 01/13/2021] [Accepted: 01/17/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Patients with relapsed small-cell lung cancer (SCLC) have few treatment options and dismal survival. Phase I/II data show activity of nivolumab in previously treated SCLC. PATIENTS AND METHODS CheckMate 331 is a randomized, open-label, phase III trial of nivolumab versus standard chemotherapy in relapsed SCLC. Patients with relapse after first-line, platinum-based chemotherapy were randomized 1 : 1 to nivolumab 240 mg every 2 weeks or chemotherapy (topotecan or amrubicin) until progression or unacceptable toxicity. Primary endpoint was overall survival (OS). RESULTS Overall, 284 patients were randomized to nivolumab and 285 to chemotherapy. Minimum follow-up was 15.8 months. No significant improvement in OS was seen with nivolumab versus chemotherapy [median OS, 7.5 versus 8.4 months; hazard ratio (HR), 0.86; 95% confidence interval (CI), 0.72-1.04; P = 0.11]. A survival benefit with nivolumab was suggested in patients with baseline lactate dehydrogenase ≤ upper limit of normal and in those without baseline liver metastases. OS (nivolumab versus chemotherapy) was similar in patients with programmed death-ligand 1 combined positive score ≥1% versus <1%. Median progression-free survival was 1.4 versus 3.8 months (HR, 1.41; 95% CI, 1.18-1.69). Objective response rate was 13.7% versus 16.5% (odds ratio, 0.80; 95% CI, 0.50-1.27); median duration of response was 8.3 versus 4.5 months. Rates of grade 3 or 4 treatment-related adverse events were 13.8% versus 73.2%. CONCLUSION Nivolumab did not improve survival versus chemotherapy in relapsed SCLC. No new safety signals were seen. In exploratory analyses, select baseline characteristics were associated with improved OS for nivolumab.
Collapse
Affiliation(s)
- D R Spigel
- Oncology Department, Sarah Cannon Research Institute/Tennessee Oncology, PLLC, Nashville, USA.
| | - D Vicente
- Department of Medical Oncology, Hosp Univ Virgen Macarena, Seville, Spain
| | - T E Ciuleanu
- Medical Oncology, Prof. Dr. Ion Chiricuta Institute of Oncology and UMF Iuliu Hatieganu, Cluj-Napoca, Romania
| | - S Gettinger
- Medical Oncology, Yale Cancer Center, New Haven, USA
| | - S Peters
- Oncology Department, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - L Horn
- Department of Medicine, Vanderbilt-Ingram Cancer Center, Nashville, USA
| | | | - N Pardo Aranda
- Thoracic Unit, Medical Oncology Department, Vall d'Hebron University Hospital and Vall d'Hebron Institute of Oncology (VHIO), Barcelona
| | - O Juan-Vidal
- Department of Medical Oncology, Hospital Universitario La Fe, Valencia, Spain
| | - Y Cheng
- Department of Thoracic Oncology, Jilin Cancer Hospital, Changchun, Jilin, China
| | - H Zhang
- Department of Oncology, Tangdu Hospital, Xi'an, Shaanxi, China
| | - M Shi
- Department of Medical Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - A Luft
- Department of Thoracic Surgery, Leningrad Regional Clinical Hospital, St. Petersburg, Russian Federation
| | - J Wolf
- Clinic I for Internal Medicine, Center for Integrated Oncology, University Hospital of Cologne, Cologne, Germany
| | - S Antonia
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, USA
| | - K Nakagawa
- Department of Medical Oncology, Kindai University Hospital, Osaka, Japan
| | - J Fairchild
- Clinical Development, Bristol Myers Squibb, Princeton, USA
| | - C Baudelet
- Global Drug Development, Biometrics & Data Sciences, Bristol Myers Squibb, Princeton, USA
| | - D Pandya
- Translational Pathology, Bristol Myers Squibb, Princeton, USA
| | - P Doshi
- Translational Medicine, Bristol Myers Squibb, Princeton, USA
| | - H Chang
- Translational Bioinformatics, Bristol Myers Squibb, Princeton, USA
| | - M Reck
- Thoracic Oncology, LungenClinic Grosshansdorf, Airway Research Center North, German Center of Lung Research, Grosshansdorf, Germany
| |
Collapse
|
147
|
Rösner E, Kaemmerer D, Neubauer E, Sänger J, Lupp A. Prognostic value of PD-L1 expression in bronchopulmonary neuroendocrine tumours. Endocr Connect 2021; 10:180-190. [PMID: 33475525 PMCID: PMC7983515 DOI: 10.1530/ec-20-0540] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 01/18/2021] [Indexed: 01/24/2023]
Abstract
Programmed death protein 1 (PD-1) and its ligand, PD-L1, have emerged as promising therapeutic targets for many types of cancer that overexpress PD-L1. However, data on PD-L1 expression levels in bronchopulmonary neuroendocrine neoplasms (BP-NEN) are limited and contradictory. In the present study, a total of 298 archived, formalin-fixed, paraffin-embedded BP-NEN samples from 97 patients diagnosed with typical carcinoid (TC), atypical carcinoid (AC), small cell lung cancer (SCLC), or large cell neuroendocrine carcinoma of the lung (LCNEC) were evaluated for PD-L1 expression by immunohistochemistry using the highly sensitive monoclonal anti-PD-L1 antibody 73-10. PD-L1 expression levels were semiquantitatively estimated by tumour grading. Of the 298 BP-NEN samples, 85% were positive for PD-L1 expression. PD-L1 immunostaining predominantly localized to the plasma membrane of both tumour cells and tumour-infiltrating immune cells. SCLC and LCNEC exhibited significantly higher PD-L1 expression levels than TC or AC. PD-L1 expression levels were also higher in patients with lymph node or distant metastases, in patients who smoked, and in patients who died during the follow-up period. Moreover, PD-L1 expression levels correlated positively with tumour grading, Ki-67 index and the expression of the chemokine receptor CXCR4 and negatively with the levels of somatostatin receptor 1 and chromogranin A. High tumour PD-L1 levels were associated with poor patient outcomes. In conclusion, PD-L1 expression is common in BP-NEN, increases with malignancy, and is associated with poor prognosis. Therefore, targeting the PD-1/PD-L1 axis could be a promising strategy for treating BP-NEN. PD-L1 may also represent a useful prognostic biomarker for this tumour entity.
Collapse
Affiliation(s)
- Erik Rösner
- Institute of Pharmacology and Toxicology, Jena University Hospital, Jena, Germany
| | - Daniel Kaemmerer
- Department of General and Visceral Surgery, Zentralklinik Bad Berka, Bad Berka, Germany
| | - Elisa Neubauer
- Institute of Pharmacology and Toxicology, Jena University Hospital, Jena, Germany
| | - Jörg Sänger
- Laboratory of Pathology and Cytology Bad Berka, Bad Berka, Germany
| | - Amelie Lupp
- Institute of Pharmacology and Toxicology, Jena University Hospital, Jena, Germany
- Correspondence should be addressed to A Lupp:
| |
Collapse
|
148
|
Winther-Larsen A, Aggerholm-Pedersen N, Sandfeld-Paulsen B. Inflammation scores as prognostic biomarkers in small cell lung cancer: a systematic review and meta-analysis. Syst Rev 2021; 10:40. [PMID: 33509254 PMCID: PMC7844954 DOI: 10.1186/s13643-021-01585-w] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 01/06/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Inflammation scores based on general inflammation markers as leucocyte count or C-reactive protein have been evaluated as prognostic markers of inferior survival in several cancers. In small cell lung cancer (SCLC), however, inflammation scores are less studied. In the present study, we set out to perform a systematic review and meta-analysis investigating reported associations between inflammation scores and overall survival (OS) in SCLC. METHODS A literature search was performed in PubMed, Embase, Scopus, and Web of Science following the Preferred Reporting Items for Systematic and Meta-Analyses (PRISMA) guidelines. Of the identified publications, only studies in English containing original data evaluating inflammation scores as a prognostic factor in SCLC patients were included. Hazard ratios (HRs) for OS were pooled in a random-effects model. RESULTS In total, 33 articles were included evaluating eight different inflammation scores in 7762 SCLC patients. Seven of the identified scores were based on leucocyte count. Neutrophil-to-lymphocyte ratio (NLR) and platelet-to-lymphocyte (PLR) ratio were the most frequently evaluated scores (NLR: n = 23; PLR: n = 22). For NLR, a meta-analysis including 16 studies demonstrated that patients with a high NLR had a significantly shorter OS compared to patients with a low NLR (pooled HR = 1.39 (95% CI, 1.23-1.56)). For PLR, an association with survival could not be confirmed in a meta-analysis performed based on eight studies (pooled HR = 1.20 (95% CI, 0.96-1.51)). CONCLUSIONS This review identifies that inflammation scores based on general inflammation markers have some potential as prognostic biomarkers in SCLC. The meta-analyses indicated that NLR is associated with inferior OS, whereas an association between PLR and OS could not be confirmed. Thus, NLR could be a useful biomarker of OS in SCLC patients. SYSTEMATIC REVIEW REGISTRATION The protocol for the study was submitted to the PROSPERO database (registration number CRD42020188553 ).
Collapse
Affiliation(s)
- Anne Winther-Larsen
- Department of Clinical Biochemistry, Viborg Regional Hospital, Viborg, Denmark
| | | | - Birgitte Sandfeld-Paulsen
- Department of Clinical Biochemistry, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200, Aarhus N, Denmark.
| |
Collapse
|
149
|
Wu J, Zhang A, Li L, Liu S, Yang F, Yang R. Meta-analysis of the Efficacy and Tolerability of Immune Checkpoint Inhibitors Combined With Chemotherapy in First-line Treatment of Small Cell Lung Cancer. Clin Ther 2021; 43:582-593.e2. [PMID: 33509647 DOI: 10.1016/j.clinthera.2020.12.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/25/2020] [Accepted: 12/25/2020] [Indexed: 12/16/2022]
Abstract
PURPOSE The present study was conducted to evaluate the efficacy and tolerability of using an immune checkpoint inhibitor (ICI; programmed cell death protein 1/ligand 1 [PD-1/PD-L1] inhibitor or cytotoxic T-lymphocyte antigen [CTLA]-4 inhibitor) combined with chemotherapy in the first-line treatment of small cell lung cancer. METHODS Potential articles and studies were identified using Web of Science, Cochrane Library, and ClinicalTrials.gov. The end points included overall survival, progression-free survival, objective response rate, and adverse events. Significant heterogeneity was represented by a P value (Ph) of <0.05 or an I2 value of ≥50%, and the random-effects model was applied for pooled analysis. Otherwise, the fixed-effects model was used. Subgroup analysis was performed based on the type of ICI. Potential publication bias was evaluated via funnel plot and the Egger test. FINDINGS Five eligible articles were included. Both overall survival (hazard ratio [HR] = 0.83; 95% CI, 0.75-0.91; P < 0.001) and progression-free survival (HR = 0.80; 95% CI, 0.73-0.86; P < 0.001) were significantly prolonged by joint ICI + chemotherapy treatment. Additionally, the rates of tolerable grade ≥3 adverse events were similar between the ICI combination regimens and conventional chemotherapy (relative risk = 1.05; 95% CI, 0.98-1.12; P = 0.17). Subanalysis demonstrated that patient survival and objective response rate were more efficiently improved with a combination of anti-PD-1/PD-L1, but not anti-CTLA-4, + chemotherapy. IMPLICATIONS Based on data from the available literature, clinical efficacy (as measured by patient survival and objective response rate) was improved with a combination of anti-PD-1/PD-L1 + chemotherapy as first-line treatment compared with chemotherapy alone in patients with small cell lung cancer.
Collapse
Affiliation(s)
- Jiao Wu
- Department of the Second Medical Oncology, Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Aifen Zhang
- Department of Gastroenterology, The Second People's Hospital of Qujing, Qujing, China
| | - Lu Li
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Sicheng Liu
- Department of the Second Medical Oncology, Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Fang Yang
- Department of the Second Medical Oncology, Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Runxiang Yang
- Department of the Second Medical Oncology, Third Affiliated Hospital of Kunming Medical University, Kunming, China.
| |
Collapse
|
150
|
Takagi H, Zhao S, Muto S, Yokouchi H, Nishihara H, Harada T, Yamaguchi H, Mine H, Watanabe M, Ozaki Y, Inoue T, Yamaura T, Fukuhara M, Okabe N, Matsumura Y, Hasegawa T, Osugi J, Hoshino M, Higuchi M, Shio Y, Kanno R, Aoki M, Tan C, Shimoyama S, Yamazaki S, Kikuchi H, Sakakibara-Konishi J, Oizumi S, Harada M, Akie K, Sugaya F, Fujita Y, Takamura K, Kojima T, Honjo O, Minami Y, Nishimura M, Dosaka-Akita H, Nakamura K, Inano A, Isobe H, Suzuki H. Delta-like 1 homolog (DLK1) as a possible therapeutic target and its application to radioimmunotherapy using 125I-labelled anti-DLK1 antibody in lung cancer models (HOT1801 and FIGHT004). Lung Cancer 2021; 153:134-142. [PMID: 33508526 DOI: 10.1016/j.lungcan.2021.01.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 12/17/2020] [Accepted: 01/10/2021] [Indexed: 02/06/2023]
Abstract
OBJECTIVES Delta-like 1 homolog (DLK1) is a non-canonical Notch ligand known to be expressed in several cancers but whose role in lung cancer is not yet fully understood. We sought to confirm DLK1 expression in small-cell lung cancer (SCLC) and non-small-cell lung cancer (NSCLC), and to examine DLK1's clinical significance. Furthermore, we examined the possible utility of DLK1 as a novel target in radioimmunotherapy (RIT). METHODS We retrospectively assessed the correlation between clinical features and DLK1 expression by immunohistochemistry in resected specimens from 112 patients with SCLC and 101 patients with NSCLC. Moreover, we performed cell and animal experiments, and examined the possibility of RIT targeting DLK1 in SCLC using iodine-125 (125I) -labeled anti-DLK1 antibody, knowing that 125I can be replaced with the alpha-particle-emitter astatine-211 (211At). RESULTS In SCLC and NSCLC, 20.5 % (23/112) and 16.8 % (17/101) of patients (respectively) had DLK1-positive tumors. In NSCLC, DLK1 expression was associated with recurrence-free survival (P < 0.01) but not with overall survival. In SCLC, there was no association between DLK1 expression and survival. In addition, 125I-labeled anti-DLK1 antibody specifically targeted DLK1 on human SCLC tumor cell lines. Furthermore, 125I-labeled anti-DLK1 antibody was incorporated into tumor tissue in a mouse model. CONCLUSION A proportion of SCLC and NSCLC exhibits DLK1 expression. As a clinical feature, DLK1 expression could be a promising prognostic factor for recurrence in patients with resected NSCLC. In addition, DLK1 could serve as a new therapeutic target, including RIT, as suggested by our pilot study using a radiolabeled anti-DLK1 antibody in SCLC.
Collapse
Affiliation(s)
- Hironori Takagi
- Department of Chest Surgery, Fukushima Medical University, Fukushima, Japan
| | - Songji Zhao
- Advanced Clinical Research Center, Fukushima Medical University, Fukushima, Japan
| | - Satoshi Muto
- Department of Chest Surgery, Fukushima Medical University, Fukushima, Japan
| | - Hiroshi Yokouchi
- Department of Respiratory Medicine, National Hospital Organization Hokkaido Cancer Center, Sapporo, Japan
| | - Hiroshi Nishihara
- Department of Translational Pathology, Hokkaido University Graduate School of Medicine, Sapporo, Japan; Genomics Unit, Keio Cancer Center, Keio University School of Medicine, Tokyo, Japan
| | - Toshiyuki Harada
- Center for Respiratory Diseases, JCHO Hokkaido Hospital, Sapporo, Japan
| | - Hikaru Yamaguchi
- Department of Chest Surgery, Fukushima Medical University, Fukushima, Japan
| | - Hayato Mine
- Department of Chest Surgery, Fukushima Medical University, Fukushima, Japan
| | - Masayuki Watanabe
- Department of Chest Surgery, Fukushima Medical University, Fukushima, Japan
| | - Yuki Ozaki
- Department of Chest Surgery, Fukushima Medical University, Fukushima, Japan
| | - Takuya Inoue
- Department of Chest Surgery, Fukushima Medical University, Fukushima, Japan
| | - Takumi Yamaura
- Department of Chest Surgery, Fukushima Medical University, Fukushima, Japan
| | - Mitsuro Fukuhara
- Department of Chest Surgery, Fukushima Medical University, Fukushima, Japan
| | - Naoyuki Okabe
- Department of Chest Surgery, Fukushima Medical University, Fukushima, Japan
| | - Yuki Matsumura
- Department of Chest Surgery, Fukushima Medical University, Fukushima, Japan
| | - Takeo Hasegawa
- Department of Chest Surgery, Fukushima Medical University, Fukushima, Japan
| | - Jun Osugi
- Department of Chest Surgery, Fukushima Medical University, Fukushima, Japan
| | - Mika Hoshino
- Department of Chest Surgery, Fukushima Medical University, Fukushima, Japan
| | - Mitsunori Higuchi
- Department of Chest Surgery, Fukushima Medical University, Fukushima, Japan
| | - Yutaka Shio
- Department of Chest Surgery, Fukushima Medical University, Fukushima, Japan
| | - Ryuzo Kanno
- Department of Thoracic Surgery, Fukushima Red Cross Hospital, Fukushima, Japan
| | - Miho Aoki
- Advanced Clinical Research Center, Fukushima Medical University, Fukushima, Japan
| | - Chengbo Tan
- Advanced Clinical Research Center, Fukushima Medical University, Fukushima, Japan
| | - Saki Shimoyama
- Advanced Clinical Research Center, Fukushima Medical University, Fukushima, Japan
| | - Shigeo Yamazaki
- Department of Thoracic Surgery, Keiyukai Sapporo Hospital, Sapporo, Japan
| | - Hajime Kikuchi
- First Department of Medicine, Hokkaido University Hospital, Sapporo, Japan; Department of Respiratory Medicine, Obihiro Kosei Hospital, Obihiro, Japan
| | | | - Satoshi Oizumi
- Department of Respiratory Medicine, National Hospital Organization Hokkaido Cancer Center, Sapporo, Japan
| | - Masao Harada
- Department of Respiratory Medicine, National Hospital Organization Hokkaido Cancer Center, Sapporo, Japan
| | - Kenji Akie
- Department of Respiratory Disease, Sapporo City General Hospital, Sapporo, Japan
| | - Fumiko Sugaya
- Department of Respiratory Medicine, Teine Keijinkai Hospital, Sapporo, Japan
| | - Yuka Fujita
- Department of Respiratory Medicine, National Hospital Organization Asahikawa Medical Center, Asahikawa, Japan
| | - Kei Takamura
- Department of Respiratory Medicine, Obihiro Kosei Hospital, Obihiro, Japan
| | - Tetsuya Kojima
- Department of Medical Oncology, KKR Sapporo Medical Center, Sapporo, Japan
| | - Osamu Honjo
- Department of Respiratory Medicine, Sapporo-Kosei General Hospital, Sapporo, Japan; Department of Respiratory Medicine, Sapporo Minami Sanjo Hospital, Sapporo, Japan
| | - Yoshinori Minami
- Respiratory Center, Asahikawa Medical University, Asahikawa, Japan
| | - Masaharu Nishimura
- First Department of Medicine, Hokkaido University Hospital, Sapporo, Japan
| | - Hirotoshi Dosaka-Akita
- Department of Medical Oncology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | | | - Akihiro Inano
- Advanced Clinical Research Center, Fukushima Medical University, Fukushima, Japan
| | - Hiroshi Isobe
- Department of Medical Oncology, KKR Sapporo Medical Center, Sapporo, Japan
| | - Hiroyuki Suzuki
- Department of Chest Surgery, Fukushima Medical University, Fukushima, Japan.
| |
Collapse
|