1
|
Ren Z, Shang S, Chen D. Recent advances in immunotherapy for small cell lung cancer. Curr Opin Oncol 2024:00001622-990000000-00220. [PMID: 39526685 DOI: 10.1097/cco.0000000000001105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
PURPOSE OF REVIEW This review aims to provide an overview of recent advances in immunotherapy for small cell lung cancer (SCLC), with a focus on the current status of immune checkpoint inhibitors (ICIs), novel combination strategies, and key biomarkers. RECENT FINDINGS The integration of ICIs into standard chemotherapy has established them as the first-line treatment for extensive-stage SCLC (ES-SCLC). The ADRIATIC trial further demonstrated the efficacy of ICI maintenance therapy in limited-stage SCLC. Additionally, combining radiotherapy with ICIs has shown promising synergistic effects, including the abscopal and radscopal effects. Ongoing investigations into the combination of ICIs with targeted therapies, such as antiangiogenic agents and DNA damage response inhibitors, have yielded encouraging preliminary results. Notably, the novel therapeutic agent tarlatamab, the first bispecific DLL3-directed CD3 T-cell engager, has recently received FDA approval for second-line treatment of ES-SCLC. Advances in omics technologies have shed light on the intra-tumor and inter-tumor heterogeneity of SCLC, leading to the identification of new molecular subtypes and biomarkers, thereby paving the way for precision medicine. SUMMARY Despite the improved outcomes associated with immunotherapy in SCLC, the overall clinical benefit remains modest. Further preclinical and clinical studies are essential to identify optimal treatment regimens and enhance therapeutic efficacy.
Collapse
Affiliation(s)
- Ziyuan Ren
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | | | | |
Collapse
|
2
|
Qiang M, Liu H, Yang L, Wang H, Guo R. Immunotherapy for small cell lung cancer: the current state and future trajectories. Discov Oncol 2024; 15:355. [PMID: 39152301 PMCID: PMC11329494 DOI: 10.1007/s12672-024-01119-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 06/21/2024] [Indexed: 08/19/2024] Open
Abstract
Small cell lung cancer (SCLC) constitutes approximately 10% to 15% of all lung cancer diagnoses and represents a pressing global public health challenge due to its high mortality rates. The efficacy of conventional treatments for SCLC is suboptimal, characterized by limited anti-tumoral effects and frequent relapses. In this context, emerging research has pivoted towards immunotherapy combined with chemotherapy, a rapidly advancing field that has shown promise in ameliorating the clinical outcomes of SCLC patients. Through originally developed for non-small cell lung cancer (NSCLC), these therapies have extended new treatment avenues for SCLC. Currently, a nexus of emerging hot-spot treatments has demonstrated significant therapeutic efficacy. Based on the amalgamation of chemotherapy and immunotherapy, and the development of new immunotherapy agents, the treatment of SCLC has seen the hoping future. Progress has been achieved in enhancing the tumor immune microenvironment through the concomitant use of chemotherapy, immunotherapy, and tyrosine kinase inhibitors (TKI), as evinced by emerging clinical trial data. Moreover, a tripartite approach involving immunotherapy, targeted therapy, and chemotherapy appears auspicious for future clinical applications. Overcoming resistance to post-immunotherapy regimens remains an urgent area of exploration. Finally, bispecific antibodies, adoptive cell transfer (ACT), oncolytic virus, monotherapy, including Delta-like ligand 3 (DLL3) and T cell immunoreceptor with Ig and ITIM domains (TIGIT), as well as precision medicine, may present a prospective route towards achieving curative outcomes in SCLC. This review aims to synthesize extant literature and highlight future directions in SCLC treatment, acknowledging the persistent challenges in the field. Furthermore, the continual development of novel therapeutic agents and technologies renders the future of SCLC treatment increasingly optimistic.
Collapse
Affiliation(s)
- Min Qiang
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Hongyang Liu
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Lei Yang
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Hong Wang
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Rui Guo
- Clinical Laboratory, The First Hospital of Jilin University, Jilin University, Changchun, China.
| |
Collapse
|
3
|
Yamamoto K, Ninomaru T, Okada H, Hirano K, Shimada T, Hata A. Continuous immunotherapy beyond progression in clinical practice for small cell lung cancer. Thorac Cancer 2024; 15:1271-1275. [PMID: 38623812 PMCID: PMC11128369 DOI: 10.1111/1759-7714.15308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 03/08/2024] [Accepted: 03/31/2024] [Indexed: 04/17/2024] Open
Abstract
In non-small-cell lung cancer, continuous immune-checkpoint inhibitors (ICIs) beyond progression are often used in clinical practice. On the other hand, there is almost no data on whether the concept of continuous ICIs beyond progression can be adopted in small-cell lung cancer (SCLC). We describe the effectiveness of continuous ICIs beyond progression in SCLC. Medical courses of SCLC patients treated with chemo-immunotherapy were retrospectively reviewed at our hospital. The study included 36 patients with a median age of 73 years (range 46-83 years) who introduced chemo-immunotherapy between September 2019 and December 2022. Atezolizumab and durvalumab in combination with platinum plus etoposide were administered in 24 and 12 patients, respectively. The overall response rate was 67% and the disease control rate was 86%. The median progression-free survival and time to treatment failure (TTF) were 5.1 and 10.3 months, respectively. The median cycle of ICIs was 5 (range 1-42). The median overall survival was 13.6 months. ICIs were administered beyond progression in 14 (39%) patients: five were treated again with chemo-immunotherapy and local ablative radiotherapy, four with local ablative radiotherapy and continuous ICIs, three with chemo-immunotherapy, and two with continuous ICIs alone. TTF exceeded 12 months in 12 (86%) of the 14 cases, six of which were still on ICIs. Adverse events ≥grade 3 were observed in 21 (58%) patients. A notable TTF suggested a benefit of continuous ICIs beyond progression. The concept could be suitably adopted and provide a favorable prognosis in selected cases of SCLC that were previously regarded as an aggressive malignancy.
Collapse
Affiliation(s)
- Ken Yamamoto
- Division of Thoracic OncologyKobe Minimally Invasive Cancer CenterKobeJapan
| | - Taira Ninomaru
- Division of Thoracic OncologyKobe Minimally Invasive Cancer CenterKobeJapan
| | - Hideaki Okada
- Division of Thoracic OncologyKobe Minimally Invasive Cancer CenterKobeJapan
| | - Katsuya Hirano
- Division of Thoracic OncologyKobe Minimally Invasive Cancer CenterKobeJapan
| | - Temiko Shimada
- Division of Thoracic OncologyKobe Minimally Invasive Cancer CenterKobeJapan
| | - Akito Hata
- Division of Thoracic OncologyKobe Minimally Invasive Cancer CenterKobeJapan
| |
Collapse
|
4
|
Benoit A, Vogin G, Duhem C, Berchem G, Janji B. Lighting Up the Fire in the Microenvironment of Cold Tumors: A Major Challenge to Improve Cancer Immunotherapy. Cells 2023; 12:1787. [PMID: 37443821 PMCID: PMC10341162 DOI: 10.3390/cells12131787] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 06/30/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023] Open
Abstract
Immunotherapy includes immune checkpoint inhibitors (ICI) such as antibodies targeting cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) or the programmed cell death protein/programmed death ligand 1 (PD-1/PD-L1) axis. Experimental and clinical evidence show that immunotherapy based on immune checkpoint inhibitors (ICI) provides long-term survival benefits to cancer patients in whom other conventional therapies have failed. However, only a minority of patients show high clinical benefits via the use of ICI alone. One of the major factors limiting the clinical benefits to ICI can be attributed to the lack of immune cell infiltration within the tumor microenvironment. Such tumors are classified as "cold/warm" or an immune "desert"; those displaying significant infiltration are considered "hot" or inflamed. This review will provide a brief summary of different tumor properties contributing to the establishment of cold tumors and describe major strategies that could reprogram non-inflamed cold tumors into inflamed hot tumors. More particularly, we will describe how targeting hypoxia can induce metabolic reprogramming that results in improving and extending the benefit of ICI.
Collapse
Affiliation(s)
- Alice Benoit
- Tumor Immunotherapy and Microenvironment (TIME) Group, Department of Cancer Research, Luxembourg Institute of Health (LIH), L-1210 Luxembourg, Luxembourg; (A.B.); (G.B.)
| | - Guillaume Vogin
- Centre National de Radiothérapie François Baclesse, L-4005 Esch-sur-Alzette, Luxembourg;
- Ingénierie Moléculaire et Physiopathologie Articulaire (IMoPA), Université de Lorraine—UMR 7365, 54505 Vandoeuvre-lès-Nancy, France
| | - Caroline Duhem
- Department of Hemato-Oncology, Centre Hospitalier du Luxembourg, L-1210 Luxembourg, Luxembourg;
| | - Guy Berchem
- Tumor Immunotherapy and Microenvironment (TIME) Group, Department of Cancer Research, Luxembourg Institute of Health (LIH), L-1210 Luxembourg, Luxembourg; (A.B.); (G.B.)
- Department of Hemato-Oncology, Centre Hospitalier du Luxembourg, L-1210 Luxembourg, Luxembourg;
- Faculty of Science, Technology and Medicine, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Bassam Janji
- Tumor Immunotherapy and Microenvironment (TIME) Group, Department of Cancer Research, Luxembourg Institute of Health (LIH), L-1210 Luxembourg, Luxembourg; (A.B.); (G.B.)
| |
Collapse
|
5
|
Tomaciello M, Conte M, Montinaro FR, Sabatini A, Cunicella G, Di Giammarco F, Tini P, Gravina GL, Cortesi E, Minniti G, De Vincentis G, Frantellizzi V, Marampon F. Abscopal Effect on Bone Metastases from Solid Tumors: A Systematic Review and Retrospective Analysis of Challenge within a Challenge. Biomedicines 2023; 11:biomedicines11041157. [PMID: 37189775 DOI: 10.3390/biomedicines11041157] [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: 03/03/2023] [Revised: 03/29/2023] [Accepted: 04/07/2023] [Indexed: 05/17/2023] Open
Abstract
BACKGROUND Abscopal effect (AE) describes the ability of radiotherapy (RT) to induce immune-mediated responses in nonirradiated distant metastasis. Bone represents the third most frequent site of metastasis and an immunologically favorable environment for the proliferation of cancer cells. We revised the literature, searching documented cases of AE involving bone metastases (BMs) and evaluated the incidence of AE involving BMs in patients requiring palliative RT on BMs or non-BMs treated at our department. METHODS Articles published in the PubMed/MEDLINE database were selected using the following search criteria: ((abscopal effect)) AND ((metastases)). Patients with BMs, who underwent performed bone scintigraphy before and at least 2-3 months after RT, were selected and screened between January 2015 and July 2022. AE was defined as an objective response according to the scan bone index for at least one nonirradiated metastasis at a distance > 10 cm from the irradiated lesion. The primary endpoint was the rate of AE on BMs. RESULTS Ten cases experiencing AE of BMs were identified from the literature and eight among our patients. CONCLUSIONS The analysis performed here suggests the use of hypofractionated radiotherapy as the only triggering factor for AE of BMs through the activation of the immune response.
Collapse
Affiliation(s)
- Miriam Tomaciello
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Division of Radiotherapy, Sapienza University of Rome, 00161 Rome, Italy
| | - Miriam Conte
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Division of Nuclear Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Francesca Romana Montinaro
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Division of Radiotherapy, Sapienza University of Rome, 00161 Rome, Italy
| | - Arianna Sabatini
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Division of Oncology, Sapienza University of Rome, 00161 Rome, Italy
| | - Giorgia Cunicella
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Division of Radiotherapy, Sapienza University of Rome, 00161 Rome, Italy
| | - Federico Di Giammarco
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Division of Radiotherapy, Sapienza University of Rome, 00161 Rome, Italy
| | - Paolo Tini
- Radiation Oncology Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy
| | - Giovanni Luca Gravina
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Enrico Cortesi
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Division of Oncology, Sapienza University of Rome, 00161 Rome, Italy
| | - Giuseppe Minniti
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Division of Radiotherapy, Sapienza University of Rome, 00161 Rome, Italy
- IRCCS Neuromed, 86077 Pozzilli, Italy
| | - Giuseppe De Vincentis
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Division of Nuclear Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Viviana Frantellizzi
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Division of Nuclear Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Francesco Marampon
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Division of Radiotherapy, Sapienza University of Rome, 00161 Rome, Italy
| |
Collapse
|
6
|
Liu MH, Li YX, Liu Z. Envafolimab combined with chemotherapy in the treatment of combined small cell lung cancer: A case report. World J Clin Cases 2023; 11:1115-1121. [PMID: 36874434 PMCID: PMC9979283 DOI: 10.12998/wjcc.v11.i5.1115] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 01/08/2023] [Accepted: 01/20/2023] [Indexed: 02/14/2023] Open
Abstract
BACKGROUND Combined small cell lung cancer (C-SCLC) is a special subtype of small cell lung cancer that is relatively rare, aggressive, and prone to early metastasis and has a poor prognosis. Currently, there are limited studies on C-SCLC, and there is no uniform standard treatment, especially for extensive C-SCLC, which still faces great challenges. In recent years, the development and progress of immunotherapy have provided more possibilities for the treatment of C-SCLC. We used immunotherapy combined with first-line chemotherapy to treat extensive-stage C-SCLC to explore its antitumor activity and safety.
CASE SUMMARY We report a case of C-SCLC that presented early with adrenal, rib, and mediastinal lymph node metastases. The patient received carboplatin and etoposide with concurrent initiation of envafolimab. After 6 cycles of chemotherapy, the lung lesion was significantly reduced, and the comprehensive efficacy evaluation showed a partial response. No serious drug-related adverse events occurred during the treatment, and the drug regimen was well tolerated.
CONCLUSION Envafolimab combined with carboplatin and etoposide in the treatment of extensive-stage C-SCLC has preliminary antitumor activity and good safety and tolerability.
Collapse
Affiliation(s)
- Mei-Hong Liu
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Dalian Medical University, Dalian 116011, Liaoning Province, China
| | - Yan-Xia Li
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Dalian Medical University, Dalian 116011, Liaoning Province, China
| | - Zhuo Liu
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Dalian Medical University, Dalian 116011, Liaoning Province, China
| |
Collapse
|