1
|
Murray NP. Immunomodulation and Immunotherapy for Patients with Prostate Cancer: An Up-to-Date Review. Biomedicines 2025; 13:1179. [PMID: 40427006 PMCID: PMC12109314 DOI: 10.3390/biomedicines13051179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 05/07/2025] [Accepted: 05/08/2025] [Indexed: 05/29/2025] Open
Abstract
Immunotherapy alone or in combination with chemotherapy or radiotherapy is the frontline treatment for melanoma and lung cancer. However, its role in prostate cancer is usually as a fourth-line treatment. It is usually employed in patients with metastasis, after androgen blockade and chemotherapy. This article reviews the immunosuppressive effects of prostate cancer and possible uses of various types of immunotherapies. It also considers when would be the optimal time to employ this type of therapy.
Collapse
Affiliation(s)
- Nigel P. Murray
- Faculty of Medicine, Universidad Finis Terrae, Santiago 7501015, Chile;
- Department of Medicine, Hospital de Carabineros de Chile, Santiago 7770199, Chile
| |
Collapse
|
2
|
Mungalov RV, Mushenkova NV, Chudakov DM, Turchaninova MA. Engaging T cells for cleanup. Front Immunol 2025; 16:1551424. [PMID: 40416957 PMCID: PMC12099299 DOI: 10.3389/fimmu.2025.1551424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Accepted: 04/07/2025] [Indexed: 05/27/2025] Open
Abstract
T-cell engagers represent a transformative approach to cancer immunotherapy leveraging bispecific and multispecific antibody constructs to redirect T-cell cytotoxicity toward malignant cells. These molecules bridge T cells and tumor cells by simultaneously binding CD3 on T cells and tumor-associated antigens on cancer cells, thereby enabling precise immune targeting even in immunologically "cold" tumors. Recent advancements include conditional T-cell engagers activated by tumor microenvironment proteases to minimize off-tumor toxicity as well as T-cell receptor-based engagers targeting intracellular antigens via MHC presentation. Clinical successes, such as Kimmtrak in metastatic uveal melanoma, underscore good potential of these modalities, while challenges persist in the management of cytokine release syndrome, neurotoxicity, and tumor resistance. Emerging multispecific engagers are aimed at enhancing efficacy via incorporation of costimulatory signals, thus offering a promising trajectory for next-generation immunotherapies. T-cell engagers are also gaining attention in the treatment of autoimmune disorders, where they can be designed to selectively modulate pathogenic immune responses. By targeting autoreactive T or B cells, T-cell engagers hold promise for restoring immune tolerance in such conditions as HLA-B*27-associated autoimmunity subtypes, multiple sclerosis, rheumatoid arthritis, and type 1 diabetes mellitus. Engineering strategies that incorporate inhibitory receptors or tissue-specific antigens may further refine T-cell engagers' therapeutic potential in autoimmunity, by minimizing systemic immunosuppression while preserving immune homeostasis.
Collapse
Affiliation(s)
- Roman V. Mungalov
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Genomics of Adaptive Immunity Department, Institute of Bioorganic Chemistry, Moscow, Russia
- Faculty of Biology and Biotechnology, Higher School of Economics, Moscow, Russia
| | - Natalia V. Mushenkova
- Genomics of Adaptive Immunity Department, Institute of Bioorganic Chemistry, Moscow, Russia
- Unicorn Capital Partners, Moscow, Russia
| | - Dmitriy M. Chudakov
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Genomics of Adaptive Immunity Department, Institute of Bioorganic Chemistry, Moscow, Russia
- Center for Molecular and Cellular Biology, Moscow, Russia
- Department of Molecular Medicine, Central European Institute of Technology, Brno, Czechia
- Abu Dhabi Stem Cell Center, Al Muntazah, United Arab Emirates
| | - Maria A. Turchaninova
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Genomics of Adaptive Immunity Department, Institute of Bioorganic Chemistry, Moscow, Russia
| |
Collapse
|
3
|
Aksoyalp ZS, Kayki-Mutlu G, Wojnowski L, Michel MC. A year in pharmacology: new drugs approved by the US Food and Drug Administration in 2024. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:5077-5099. [PMID: 40163152 PMCID: PMC11985671 DOI: 10.1007/s00210-025-04020-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Accepted: 03/04/2025] [Indexed: 04/02/2025]
Abstract
The US Food and Drug Administration approved 50 new drugs and nine new cellular and gene therapy products in 2024, i.e., a total of 59 new medical therapies. The latter group represented three treatments each for oncology and hematology/immunotherapy, and one each for neurology, genetic disorders, and cardiovascular disorders. Oncology, hematology/immunotherapy, and neurological disorders (14, six, and seven, respectively) also were highly prevalent among classic medications. Looking at trends over the past 5 years, we observe a greater share in first-in-class medications, more fast-track approvals, and mRNA/gene/cell-based therapies. While small molecules remain the largest fraction, their percentage has been declining substantially over the past 5 years. Taking together, these findings testify to the commitment of the pharmaceutical industry for innovative treatments, including conditions for which no approved therapies existed. On the other hand, there also is a trend for approvals for narrowly focused conditions such as tumors defined by genetic alterations.
Collapse
Affiliation(s)
- Zinnet Sevval Aksoyalp
- Department of Pharmacology, Faculty of Pharmacy, Izmir Katip Celebi University, Izmir, Turkey
| | - Gizem Kayki-Mutlu
- Department of Pharmacology, Faculty of Pharmacy, Ankara University, Ankara, Turkey
| | - Leszek Wojnowski
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Martin C Michel
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University, Mainz, Germany.
| |
Collapse
|
4
|
Zhu X, Ding CKC, Aggarwal RR. Emerging Therapeutic Targets of Neuroendocrine Prostate Cancer. Curr Oncol Rep 2025; 27:362-374. [PMID: 40011325 DOI: 10.1007/s11912-025-01643-9] [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] [Accepted: 01/27/2025] [Indexed: 02/28/2025]
Abstract
PURPOSE OF REVIEW Treatment-emergent neuroendocrine prostate cancer (NEPC) is aggressive and lethal. As androgen receptor signaling inhibitors (ARSIs) are increasingly used in earlier disease settings, treatment-emergent NEPC becomes more prevalent, and effective therapies are urgently needed. The purpose of this review was to summarize recent progress on emerging therapeutic targets of NEPC. RECENT FINDINGS A multitude of therapeutic targets have emerged in NEPC over recent years. These targets may represent drivers of treatment-emergent lineage plasticity or simply be overexpressed on the surface of NEPC cells. Multiple modalities have been employed to drug these targets, with promising preclinical and clinical results. Treatment-emergent NEPC represents a distinct and clinically significant subset of castration-resistant prostate cancer (CRPC). Emerging therapeutic approaches have demonstrated encouraging efficacy and safety profiles, offering the potential to improve patient outcomes.
Collapse
Affiliation(s)
- Xiaolin Zhu
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
- Division of Hematology and Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Chien-Kuang C Ding
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
- Department of Anatomic Pathology, University of California San Francisco, San Francisco, CA, USA
| | - Rahul R Aggarwal
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA.
- Division of Hematology and Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
5
|
Malkowski KD. Taking a BiTE out of cancer. Nursing 2025; 55:46-49. [PMID: 40122873 DOI: 10.1097/nsg.0000000000000172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
ABSTRACT Tarlatamab (tarlatamab-dlle) is the first bispecific T-cell engager (BiTE) therapy approved for treating a solid tumor, specifically small cell lung cancer, making it the first of its kind. It targets delta-like ligand 3 antigens on the surface of tumor cells while activating the patient's T cells. This article discusses BiTE therapy as well as its boxed warnings and precautions that nurses need to know to provide optimum patient care.
Collapse
Affiliation(s)
- Kimberly D Malkowski
- Kimberly Malkowski is an oncology treatment nurse at UPMC Hillman Cancer Center and a member of the Nursing2025 Editorial Board
| |
Collapse
|
6
|
Chiang AC, Olmedo Garcia ME, Carlisle JW, Dowlati A, Reguart N, Felip E, Jost PJ, Steeghs N, Stec R, Gadgeel SM, Loong HH, Jiang W, Hamidi A, Parkes A, Paz-Ares L. Safety of tarlatamab with 6-8-h outpatient versus 48-h inpatient monitoring during cycle 1: DeLLphi-300 phase 1 substudy. ESMO Open 2025; 10:104538. [PMID: 40187110 PMCID: PMC12002761 DOI: 10.1016/j.esmoop.2025.104538] [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: 12/20/2024] [Revised: 02/28/2025] [Accepted: 03/06/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Tarlatamab, a bispecific T-cell engager immunotherapy targeting delta-like ligand 3, has demonstrated promising survival outcomes in small-cell lung cancer (SCLC). Given the risk of cytokine release syndrome (CRS), initial clinical trials incorporated 48-72-h inpatient monitoring in cycle 1. METHODS Patients with previously treated SCLC were enrolled into DeLLphi-300 part F, which evaluated the safety of tarlatamab 10 mg every 2 weeks (Q2W) with 6-8-h outpatient monitoring following cycle 1 doses. The primary endpoint, safety, was compared with patients from DeLLphi-300 part A receiving tarlatamab 10 mg Q2W with 48-h inpatient monitoring for cycle 1 doses. RESULTS In cycle 1, the rates of treatment-related adverse events and hospitalizations, including emergency room visits, were similar between outpatient (n = 30) and inpatient (n = 58) groups (93% versus 100% and 27% versus 34%, respectively). The incidence of all grade and serious CRS during cycle 1 was similar between outpatient and inpatient groups (any grade: 60% versus 62%; serious: 17% versus 22%). The median time to CRS resolution was 3 days for both groups. CONCLUSIONS Safety outcomes, including hospitalization rates, were similar in this first-in-human study following tarlatamab 10 mg Q2W administration with 6-8-h outpatient versus 48-h inpatient monitoring in cycle 1.
Collapse
Affiliation(s)
- A C Chiang
- Division of Thoracic Medical Oncology, Yale University School of Medicine, New Haven, USA.
| | - M E Olmedo Garcia
- Department of Medical Oncology, Ramón y Cajal University Hospital, Madrid, Spain
| | - J W Carlisle
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, USA
| | - A Dowlati
- Division of Hematology and Oncology, Department of Medicine, University Hospitals Seidman Cancer Center, Cleveland, USA; Case Western Reserve University, Cleveland, USA
| | - N Reguart
- Department of Oncology, Hospital Clinic y Provincial de Barcelona, Barcelona, Spain
| | - E Felip
- Medical Oncology Service (Lung Cancer Unit), Vall d'Hebron University Hospital, Barcelona, Spain; Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - P J Jost
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - N Steeghs
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - R Stec
- Biokinetica, Przychodnia Józefów, Józefów, Poland; Department of Oncology, Warsaw Medical University, Warsaw, Poland
| | - S M Gadgeel
- Department of Oncology, Henry Ford Cancer Institute/Henry Ford Health, Detroit, USA
| | - H H Loong
- Department of Clinical Oncology, The Chinese University of Hong Kong, Hong Kong, China; Phase 1 Clinical Trial Centre, The Chinese University of Hong Kong, Hong Kong, China
| | - W Jiang
- Amgen Inc., Thousand Oaks, USA
| | | | | | - L Paz-Ares
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain; CNIO-H12O Lung Cancer Unit, Madrid, Spain; Ciberonc, Madrid, Spain; Universidad Complutense, Madrid, Spain
| |
Collapse
|
7
|
Saadh MJ, Khidr WA, Alfarttoosi KH, Bishoyi AK, Ganesan S, Shankhyan A, Gayathri S, Rizaev J, Taher WM, Alwan M, Jawad MJ, Al-Nuaimi AMA. Metal nanoparticles as a promising therapeutic approach for prostate cancer diagnosis and therapy: a comprehensive review. Med Oncol 2025; 42:83. [PMID: 39987535 DOI: 10.1007/s12032-025-02633-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 02/13/2025] [Indexed: 02/25/2025]
Abstract
Prostate cancer is a leading cause of mortality among men worldwide, particularly in the USA and European nations, with an estimated 1.9 million new cases and over 580,000 deaths annually, according to recent global statistics. The treatment of prostate tumors presents significant clinical challenges, due to the disease's high metastatic potential, specifically to vital organs, such as the liver, lungs, bones, and brain. The intrinsic heterogeneity of prostate cancer cells, characterized by diverse genetic, molecular, and phenotypic profiles, complicates conventional therapeutic strategies, highlighting the need for advanced diagnostic and treatment modalities. Nanoparticles play a critical role in oncology field due to their unique physicochemical properties, including high surface area-to-volume ratio and the ability to be functionalized with targeting ligands. Metallic-based nanoparticles exhibits significant potential for applications in field of nanomedicine, drug delivery systems, gene silencing methods, radiotherapy enhancement, cancer diagnostics, and targeted therapeutic interventions. Metal nanoparticles have substantially improved the sensitivity and specificity of major imaging modalities and have demonstrated remarkable efficacy as biosensors for the detection of prostate cancer-specific biomarkers. This review article provides an in-depth analysis of the utilization of metal nanomaterials in prostate cancer, focusing on their roles in enhancing therapeutic efficacy, advancing diagnostic precision, and supporting the development of novel treatment strategies.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan
| | - Wajida Ataallah Khidr
- Department of Medical Laboratory Technics, College of Health and Medical Technology, Alnoor University, Mosul, Iraq.
| | | | - Ashok Kumar Bishoyi
- Department of Microbiology, Marwadi University Research Center, Faculty of Science, Marwadi University, Rajkot, Gujarat, 360003, India
| | - Subbulakshmi Ganesan
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Aman Shankhyan
- Centre for Research Impact and Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab, 140401, India
| | - S Gayathri
- Department of Chemistry, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India
| | - Jasur Rizaev
- Department of Public Health and Healthcare Management, Rector, Samarkand State Medical University, 18, Amir Temur Street, Samarkand, Uzbekistan
| | - Waam Mohammed Taher
- College of Nursing, National University of Science and Technology, Nasiriyah, Dhi Qar, Iraq
| | - Mariem Alwan
- Pharmacy College, Al-Farahidi University, Baghdad, Iraq
| | | | | |
Collapse
|
8
|
Ma M, Zhang Y, Pu K, Tang W. Nanomaterial-enabled metabolic reprogramming strategies for boosting antitumor immunity. Chem Soc Rev 2025; 54:653-714. [PMID: 39620588 DOI: 10.1039/d4cs00679h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2025]
Abstract
Immunotherapy has become a crucial strategy in cancer treatment, but its effectiveness is often constrained. Most cancer immunotherapies focus on stimulating T-cell-mediated immunity by driving the cancer-immunity cycle, which includes tumor antigen release, antigen presentation, T cell activation, infiltration, and tumor cell killing. However, metabolism reprogramming in the tumor microenvironment (TME) supports the viability of cancer cells and inhibits the function of immune cells within this cycle, presenting clinical challenges. The distinct metabolic needs of tumor cells and immune cells require precise and selective metabolic interventions to maximize therapeutic outcomes while minimizing adverse effects. Recent advances in nanotherapeutics offer a promising approach to target tumor metabolism reprogramming and enhance the cancer-immunity cycle through tailored metabolic modulation. In this review, we explore cutting-edge nanomaterial strategies for modulating tumor metabolism to improve therapeutic outcomes. We review the design principles of nanoplatforms for immunometabolic modulation, key metabolic pathways and their regulation, recent advances in targeting these pathways for the cancer-immunity cycle enhancement, and future prospects for next-generation metabolic nanomodulators in cancer immunotherapy. We expect that emerging immunometabolic modulatory nanotechnology will establish a new frontier in cancer immunotherapy in the near future.
Collapse
Affiliation(s)
- Muye Ma
- Department of Diagnostic Radiology, Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Dr, Singapore, 117597, Singapore.
| | - Yongliang Zhang
- Department of Microbiology and Immunology, Immunology Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, 5 Science Dr 2, Singapore, 117545, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, 28 Medical Dr, Singapore, 117597, Singapore
| | - Kanyi Pu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore.
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, Singapore, 636921, Singapore
| | - Wei Tang
- Department of Diagnostic Radiology, Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Dr, Singapore, 117597, Singapore.
- Department of Pharmacy and Pharmaceutic Sciences, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore, 117543, Singapore
| |
Collapse
|
9
|
Dómine Gómez M, Subbiah V, Peters S, Sala MA, Trigo J, Paz-Ares L, Nieto Archilla A, Gomez Garcia J, Alvarez García C, López-Vilariño de Ramos JA, Kahatt Lopez C, Fernandez CM. Lurbinectedin is an effective alternative to platinum rechallenge and may restore platinum sensitivity in patients with sensitive relapsed small cell lung cancer. Expert Rev Anticancer Ther 2025; 25:27-40. [PMID: 39660812 DOI: 10.1080/14737140.2024.2438067] [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: 11/11/2024] [Revised: 11/29/2024] [Accepted: 12/02/2024] [Indexed: 12/12/2024]
Abstract
INTRODUCTION Platinum rechallenge is recommended for patients with small cell lung cancer (SCLC) who relapse ≥90 days after completing first-line chemotherapy, although it may not always be the most suitable option. AREAS COVERED Articles for review were identified via PubMed and ClinicalTrials.gov searches, supplemented with non-indexed publications (e.g. conference abstracts) known to the manufacturer. We examined evidence for platinum re-exposure in patients with sensitive relapsed SCLC, and present lurbinectedin as a potential alternative. The complementary mechanisms of action of lurbinectedin and platinum, owing to opposite sensitivity of SCLC cells, may resensitize tumor cells to platinum. As efficacy outcomes with lurbinectedin are equivalent or better than those with platinum rechallenge and its hematological safety profile is more favorable, achieving maximum dose intensity is more likely. The simpler dosing schedule of lurbinectedin (1 vs 3 days) and lack of need for granulocyte colony-stimulating factor primary prophylaxis lessens treatment burden. EXPERT OPINION Incorporation of lurbinectedin into therapeutic algorithms for relapsed SCLC has challenged long-established treatment paradigms. Initial evidence indicates that using lurbinectedin after failure of first-line platinum may prolong the platinum-free interval and reserve platinum for later use. Current evidence supports lurbinectedin as a second-line option in patients with sensitive relapsed SCLC.
Collapse
Affiliation(s)
- Manuel Dómine Gómez
- Hospital Universitario Fundación Jiménez Díaz, IIS-FJD, Universidad Autónoma de Madrid, Madrid, Spain
| | - Vivek Subbiah
- Early-Phase Drug Development, Sarah Cannon Research Institute, Nashville, TN, USA
| | - Solange Peters
- Oncology Department - CHUV, Lausanne University, Lausanne, Switzerland
| | - María Angeles Sala
- Medical Oncology Department, Hospital Universitario Basurto, Bilbao, Spain
| | - José Trigo
- Medical Oncology Department, Hospital HC Marbella, Málaga, Spain
| | - Luis Paz-Ares
- Medical Oncology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | | | | | | | | | | | | |
Collapse
|
10
|
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
|
11
|
Roskoski R. Targeted and cytotoxic inhibitors used in the treatment of lung cancers. Pharmacol Res 2024; 209:107465. [PMID: 39426470 DOI: 10.1016/j.phrs.2024.107465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 10/11/2024] [Indexed: 10/21/2024]
Abstract
Lung cancer is the leading cause of cancer deaths in the United States and the world. It is divided into two major types: small cell lung cancer (SCLC) and non-small cell lung cancer (NSCLC). In the tumor-node-metastasis (TNM) cancer-staging classification system (Stages I/II/III/IV), the severity of neoplastic growth is characterized by the size of the tumor (T1 to T4), the extent of lymph node involvement (N0 to N3), and whether (M1) or not (M0) distant metastasis has occurred. Surgery is the treatment of choice for medically fit patients with Stage I/II NSCLC. Combination chemoradiotherapy and immune checkpoint inhibitor therapy are used across all NSCLC types. Oncogene-addicted tumors with sensitizing EGFR or BRAF mutations or activating ALK, ROS1 or NTRK translocations are treated with their cognate orally active small molecule protein kinase blockers. On the order of 20 % of NSCLCs bear activating mutations in EGFR and are treated with osimertinib and other kinase antagonists. SCLC, which accounts for approximately 15 % of lung cancer cases, is a deadly high-grade neuroendocrine carcinoma with a poor prognosis. Limited-stage SCLC is confined to one hemi-thorax and one radiation port and extensive-stage disease signifies those cancers that do not meet the criteria for limited-stage disease. Local treatment options to control thoracic disease include radiotherapy and surgery. In patients with extensive-stage disease, a platinum agent (cisplatin or carboplatin) combined with etoposide and an anti-PDL1 inhibitor (atezolizumab or durvalumab) for four cycles followed by anti-PDL1 maintenance therapy is the recommended first-line regimen.
Collapse
Affiliation(s)
- Robert Roskoski
- Blue Ridge Institute for Medical Research, 221 Haywood Knolls Drive, Hendersonville, NC 28791, United States.
| |
Collapse
|
12
|
Silva S, Sousa JC, Nogueira C, Feijo R, Neto FM, Marinho LC, Sousa G, Denninghoff V, Tavora F. Relationship between the expressions of DLL3, ASC1, TTF-1 and Ki-67: First steps of precision medicine at SCLC. Oncotarget 2024; 15:750-763. [PMID: 39392394 PMCID: PMC11468345 DOI: 10.18632/oncotarget.28660] [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: 07/29/2024] [Accepted: 09/17/2024] [Indexed: 10/12/2024] Open
Abstract
This study presents an observational, cross-sectional analysis of 64 patients diagnosed with small cell lung cancer (SCLC) at a reference laboratory for thoracic pathology between 2022 and 2024. The primary objective was to evaluate the expression of Delta-like ligand 3 (DLL3) and other neuroendocrine markers such as Chromogranin, and Synaptophysin, utilizing both traditional immunohistochemistry and digital pathology tools. Patients were primarily older adults, with a median age of over 71, and most biopsies were obtained from lung parenchyma. Immunohistochemistry (IHC) was performed using specific monoclonal antibodies, with DLL3 showing variable expression across the samples. Notably, DLL3 was expressed in 72.3% of the cases, with varied intensities and a semi-quantitative H-score applied for more nuanced analysis. ASCL1 was expressed in 97% of cases, with the majority considered low-expressors. Only 11% had high expression. TTF-1, traditionally not a conventional marker for the diagnosis of SCLC, was positive in half of the cases, suggesting its potential as a biomarker. The study underscores the significant variability in the expression of neuroendocrine markers in SCLC, with implications for both diagnosis and potential therapeutic targeting. DLL3, particularly, shows promise as a therapeutic target due to its high expression rate in the cohort. The use of digital pathology software QuPath enhanced the accuracy and depth of analysis, allowing for detailed morphometric analysis and potentially informing more personalized treatment approaches. The findings emphasize the need for further research into the role of these markers in the management and treatment of SCLC, considering the poor prognosis and high mortality rate observed in the cohort.
Collapse
Affiliation(s)
- Samuel Silva
- Department of Pathology, Faculty of Medicine, Federal University of Ceará, Fortaleza (Ceará), Brazil
- ARGOS Laboratory, Fortaleza (Ceará), Brazil
| | | | - Cleto Nogueira
- Department of Pathology, Faculty of Medicine, Federal University of Ceará, Fortaleza (Ceará), Brazil
- ARGOS Laboratory, Fortaleza (Ceará), Brazil
| | - Raquel Feijo
- Department of Pathology, Faculty of Medicine, Federal University of Ceará, Fortaleza (Ceará), Brazil
- Messejana Heart and Lung Hospital, Fortaleza (Ceará), Brazil
| | | | - Laura Cardoso Marinho
- Department of Pathology, Faculty of Medicine, Federal University of Ceará, Fortaleza (Ceará), Brazil
- ARGOS Laboratory, Fortaleza (Ceará), Brazil
| | | | - Valeria Denninghoff
- Molecular Oncology Clinical Lab, University of Buenos Aires (UBA)—National Council for Scientific and Technical Research (CONICET), Buenos Aires, Argentina
- Liquid Biopsy and Cancer Interception Unit, GENYO, Centre for Genomics and Oncological Research (Pfizer/University of Granada/Andalusian Regional Government), Granada, Spain
| | - Fabio Tavora
- Department of Pathology, Faculty of Medicine, Federal University of Ceará, Fortaleza (Ceará), Brazil
- ARGOS Laboratory, Fortaleza (Ceará), Brazil
| |
Collapse
|
13
|
Raja A, Kasana A, Verma V. Next-Generation Therapeutic Antibodies for Cancer Treatment: Advancements, Applications, and Challenges. Mol Biotechnol 2024:10.1007/s12033-024-01270-y. [PMID: 39222285 DOI: 10.1007/s12033-024-01270-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 08/24/2024] [Indexed: 09/04/2024]
Abstract
The field of cancer treatment has evolved significantly over the last decade with the emergence of next-generation therapeutic antibodies. Conventional treatments like chemotherapy pose significant challenges, including adverse side effects. Monoclonal antibodies have paved the way for more targeted and effective interventions. The evolution from chimeric to humanized and fully human antibodies has led to a reduction in immunogenicity and enhanced tolerance in vivo. The advent of next-generation antibodies, including bispecific antibodies, nanobodies, antibody-drug conjugates, glyco-engineered antibodies, and antibody fragments, represents a leap forward in cancer therapy. These innovations offer increased potency, adaptability, and reduced drug resistance. Challenges such as target validation, immunogenicity, and high production costs exist. However, technological advancements in antibody engineering techniques provide optimism for addressing these issues. The future promises a paradigm shift, where ongoing research will propel these powerful antibodies to the forefront, revolutionizing the fight against cancer and creating new preventive and curative treatments. This review provides an overview of three next-generation antibody-based molecules, namely bispecific antibodies, antibody-drug conjugates, and nanobodies that have shown promising results in cancer treatment. It discusses the evolution of antibodies from conventional forms to next-generation molecules, along with their applications in cancer treatment, production methods, and associated challenges. The review aims to offer researchers insights into the evolving landscape of next-generation antibody-based cancer therapeutics and their potential to revolutionize treatment strategies.
Collapse
Affiliation(s)
- Abhavya Raja
- Department of Biotechnology, School of Engineering and Applied Sciences, Bennett University, Greater Noida, 201310, Uttar Pradesh, India
| | - Abhishek Kasana
- Department of Biotechnology, School of Engineering and Applied Sciences, Bennett University, Greater Noida, 201310, Uttar Pradesh, India
| | - Vaishali Verma
- Department of Biotechnology, School of Engineering and Applied Sciences, Bennett University, Greater Noida, 201310, Uttar Pradesh, India.
| |
Collapse
|