1
|
Alexander M, Rogers J, Parakh S, Mitchell P, Clay TD, Kao S, Hughes BGM, Itchins M, Kong BY, Pavlakis N, Solomon BJ, John T. Lurbinectedin in small cell lung cancer: real-world experience of a multicentre national early access programme. Intern Med J 2024; 54:1087-1096. [PMID: 38369719 DOI: 10.1111/imj.16348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 01/24/2024] [Indexed: 02/20/2024]
Abstract
BACKGROUND AND AIMS Lurbinectedin is a novel oncogenic transcription inhibitor active in several cancers, including small cell lung cancer (SCLC). We aimed to describe the first Australian experience of the clinical efficacy and tolerability of lurbinectedin for the treatment of SCLC after progression on platinum-containing therapy. METHODS Multicentre real-world study of individuals with SCLC initiating lurbinectedin monotherapy (3.2 mg/m2 three-weekly) on an early access programme between May 2020 and December 2021. Key outcomes were clinical utilisation, efficacy and tolerability. Progression-free survival (PFS) and overall survival (OS) were calculated using the Kaplan-Meier method. Outcome data were collected within the AUstralian Registry and biObank of thoRacic cAncers (AURORA). RESULTS Data were analysed for 46 individuals across seven sites. Lurbinectedin was given as second- (83%, 38/46) or subsequent- (17%, 8/46) line therapy, mostly with prior chemoimmunotherapy (87%, 40/46). We report dose modifications (17%, 8/46), interruptions/delays (24%, 11/46), high-grade toxicities (28%, 13/46) and hospitalisations (54%, 25/46) during active treatment. The overall response rate was 33% and the disease control rate was 50%. Six-month OS was 44% (95% confidence interval (CI): 29.0-57.1). Twelve-month OS was 15% (95% CI: 6.5-26.8). From lurbinectedin first dose, the median PFS was 2.5 months (95% CI: 1.8-2.9) and OS was 4.5 months (95% CI: 3.5-7.2). From SCLC diagnosis, the median OS was 12.9 months (95% CI: 11.0-17.2). Individuals with a longer chemotherapy-free interval prior to lurbinectedin had longer PFS and OS. CONCLUSION This real-world national experience of lurbinectedin post-platinum chemotherapy and immunotherapy for individuals with SCLC was similar to that reported in clinical trials.
Collapse
Affiliation(s)
- Marliese Alexander
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Jennifer Rogers
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Sagun Parakh
- Austin Hospital, Olivia Newton-John Cancer Research Institute, Melbourne, Victoria, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, Victoria, Australia
| | - Paul Mitchell
- Austin Hospital, Olivia Newton-John Cancer Research Institute, Melbourne, Victoria, Australia
- Division of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Victoria, Australia
| | - Timothy D Clay
- Saint John of God Subiaco Hospital, Perth, Western Australia, Australia
- Icon Cancer Care Midland, Perth, Western Australia, Australia
- School of Medical and Health Sciences, Edith Cowan University, Perth, Western Australia, Australia
| | - Steven Kao
- Chris O'Brien Lifehouse, Sydney, New South Wales, Australia
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Brett G M Hughes
- The Prince Charles Hospital, Brisbane, Queensland, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Malinda Itchins
- Royal North Shore Hospital, Sydney, New South Wales, Australia
- Northern Clinical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Benjamin Y Kong
- Royal North Shore Hospital, Sydney, New South Wales, Australia
| | - Nick Pavlakis
- Royal North Shore Hospital, Sydney, New South Wales, Australia
- Northern Clinical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Benjamin J Solomon
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Thomas John
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
2
|
Khoury R, Assi T, Ibrahim R, Ibrahim T, Verret B, Henon C, Bahleda R, Le Cesne A. A Comprehensive Review on the Role of Lurbinectedin in Soft Tissue Sarcomas. Curr Treat Options Oncol 2024; 25:176-190. [PMID: 38324075 DOI: 10.1007/s11864-024-01178-4] [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: 01/08/2024] [Indexed: 02/08/2024]
Abstract
OPINION STATEMENT Soft tissue sarcoma (STS), a substantial group of aggressive and rare tumors with tissue heterogeneity, is infrequently represented in clinical trials with an urgent necessity for newer treatment options. Lurbinectedin, an analog of trabectedin, is currently approved, in various countries, as a single agent, for the treatment of patients with relapsed small cell lung cancer (SCLC). However, preclinical and phase I and phase II trials have demonstrated the efficacy of lurbinectedin in different tumor types, including STS. The better understanding of the pathophysiology and evolution of STS as well as the mechanism of action of lurbinectedin in addition to the available data regarding the activity of this drug in this subset of patients will pave the way to newer therapeutic options and strategies.
Collapse
Affiliation(s)
- Rita Khoury
- Division of International Patients Care, Gustave Roussy Cancer Campus, 114, Rue Edouard Vaillant, Villejuif, France
| | - Tarek Assi
- Division of International Patients Care, Gustave Roussy Cancer Campus, 114, Rue Edouard Vaillant, Villejuif, France.
- Sarcoma Unit, Gustave Roussy Cancer Campus, 114, Rue Edouard Vaillant, Villejuif, France.
| | - Rebecca Ibrahim
- Division of International Patients Care, Gustave Roussy Cancer Campus, 114, Rue Edouard Vaillant, Villejuif, France
| | - Tony Ibrahim
- Division of International Patients Care, Gustave Roussy Cancer Campus, 114, Rue Edouard Vaillant, Villejuif, France
| | - Benjamin Verret
- Sarcoma Unit, Gustave Roussy Cancer Campus, 114, Rue Edouard Vaillant, Villejuif, France
| | - Clemence Henon
- Sarcoma Unit, Gustave Roussy Cancer Campus, 114, Rue Edouard Vaillant, Villejuif, France
| | - Ratislav Bahleda
- Sarcoma Unit, Gustave Roussy Cancer Campus, 114, Rue Edouard Vaillant, Villejuif, France
| | - Axel Le Cesne
- Division of International Patients Care, Gustave Roussy Cancer Campus, 114, Rue Edouard Vaillant, Villejuif, France
- Sarcoma Unit, Gustave Roussy Cancer Campus, 114, Rue Edouard Vaillant, Villejuif, France
| |
Collapse
|
3
|
Canova S, Trevisan B, Abbate MI, Colonese F, Sala L, Baggi A, Bianchi SP, D'Agostino A, Cortinovis DL. Novel Therapeutic Options for Small Cell Lung Cancer. Curr Oncol Rep 2023; 25:1277-1294. [PMID: 37870696 PMCID: PMC10640463 DOI: 10.1007/s11912-023-01465-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/19/2023] [Indexed: 10/24/2023]
Abstract
PURPOSE OF REVIEW The aim of this review is to focus on the recent advances in the molecular knowledge of small cell lung cancer (SCLC) and potential promising new treatment strategies, like targeting the DNA damage pathway, epigenetics, angiogenesis, and oncogenic drivers. RECENT FINDINGS In the last few years, the addition of immunotherapy to chemotherapy has led to significant improvements in clinical outcomes in this complex neoplasia. Nevertheless, the prognosis remains dismal. Recently, numerous genomic alterations have been identified, and they may be useful to classify SCLC into different molecular subtypes (SCLC-A, SCLC-I, SCLC-Y, SCLC-P). SCLC accounts for 10-20% of all lung cancers, most patients have an extensive disease at the diagnosis, and it is characterized by poor prognosis. Despite the progresses in the knowledge of the disease, efficacious targeted treatments are still lacking. In the near future, the molecular characterisation of SCLC will be fundamental to find more effective treatment strategies.
Collapse
Affiliation(s)
- Stefania Canova
- SC Medical Oncology, Fondazione IRCCS San Gerardo Dei Tintori, Monza, Italy
| | - Benedetta Trevisan
- SC Medical Oncology, Fondazione IRCCS San Gerardo Dei Tintori, Monza, Italy
- Department of Medical-Surgical Specialties, University of Brescia, Radiological Sciences and Public Health, Brescia, Italy
| | - Maria Ida Abbate
- SC Medical Oncology, Fondazione IRCCS San Gerardo Dei Tintori, Monza, Italy
| | - Francesca Colonese
- SC Medical Oncology, Fondazione IRCCS San Gerardo Dei Tintori, Monza, Italy
| | - Luca Sala
- SC Medical Oncology, Fondazione IRCCS San Gerardo Dei Tintori, Monza, Italy
| | - Alice Baggi
- SC Medical Oncology, Fondazione IRCCS San Gerardo Dei Tintori, Monza, Italy
- Department of Medical-Surgical Specialties, University of Brescia, Radiological Sciences and Public Health, Brescia, Italy
| | - Sofia Paola Bianchi
- Radiation Oncology Department, Fondazione IRCCS San Gerardo Dei Tintori, Monza, Italy
- School of Medicine and Surgery, University of Milano Bicocca, Milan, Italy
| | - Anna D'Agostino
- SC Medical Oncology, Fondazione IRCCS San Gerardo Dei Tintori, Monza, Italy
| | - Diego Luigi Cortinovis
- SC Medical Oncology, Fondazione IRCCS San Gerardo Dei Tintori, Monza, Italy.
- Medicine and Surgery Department, University of Milano Bicocca, Milan, Italy.
| |
Collapse
|
4
|
Cani M, Napoli VM, Garbo E, Ferrari G, Del Rio B, Novello S, Passiglia F. Targeted Therapies in Small Cell Lung Cancer: From Old Failures to Novel Therapeutic Strategies. Int J Mol Sci 2023; 24:ijms24108883. [PMID: 37240229 DOI: 10.3390/ijms24108883] [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: 05/02/2023] [Revised: 05/13/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023] Open
Abstract
The clinical management of small cell lung cancer (SCLC) treatment remains a major challenge for thoracic oncologists, with very few therapeutic advances significantly impacting patients' survival. The recent introduction of immunotherapy in the clinical setting produced a marginal benefit for a limited subset of metastatic patients, while the therapeutic scenario for relapsing extended-disease small cell lung cancers (ED-SCLCs) remains almost deserted. Recent efforts clarified the molecular features of this disease, leading to the identification of key signalling pathways which may serve as potential targets for clinical use. Despite the large number of molecules tested and the numerous therapeutic failures, some targeted therapies have recently shown interesting preliminary results. In this review, we describe the main molecular pathways involved in SCLC development/progression and provide an updated summary of the targeted therapies currently under investigation in SCLC patients.
Collapse
Affiliation(s)
- Massimiliano Cani
- Department of Oncology, University of Turin, San Luigi Hospital, 10043 Orbassano, TO, Italy
| | - Valerio Maria Napoli
- Department of Oncology, University of Turin, San Luigi Hospital, 10043 Orbassano, TO, Italy
| | - Edoardo Garbo
- Department of Oncology, University of Turin, San Luigi Hospital, 10043 Orbassano, TO, Italy
| | - Giorgia Ferrari
- Department of Oncology, University of Turin, San Luigi Hospital, 10043 Orbassano, TO, Italy
| | - Benedetta Del Rio
- Department of Oncology, University of Turin, San Luigi Hospital, 10043 Orbassano, TO, Italy
| | - Silvia Novello
- Department of Oncology, University of Turin, San Luigi Hospital, 10043 Orbassano, TO, Italy
| | - Francesco Passiglia
- Department of Oncology, University of Turin, San Luigi Hospital, 10043 Orbassano, TO, Italy
| |
Collapse
|
5
|
Moliner L, Zhang B, Lamberti G, Ardizzoni A, Byers LA, Califano R. Novel therapeutic strategies for recurrent SCLC. Crit Rev Oncol Hematol 2023; 186:104017. [PMID: 37150311 DOI: 10.1016/j.critrevonc.2023.104017] [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: 12/01/2022] [Revised: 04/25/2023] [Accepted: 05/04/2023] [Indexed: 05/09/2023] Open
Abstract
Therapeutic options for patients with relapsed SCLC are limited, and the prognosis in this setting remains poor. While clinical outcomes for frontline treatment have modestly improved with the introduction of immunotherapy, treatment in the second-line setting persists almost unchanged. In this review, current treatment options and recent advances in molecular biology are described. Emerging therapeutic options in this setting and potential strategies to improve clinical outcomes of these patients are also addressed.
Collapse
Affiliation(s)
- Laura Moliner
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, M20 4BX, UK
| | - Bingnan Zhang
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Giuseppe Lamberti
- Department of Specialized, Experimental and Diagnostic Medicine, University of Bologna, Bologna, 40138, Italy
| | - Andrea Ardizzoni
- Department of Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, 40138, Italy
| | - Lauren A Byers
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Raffaele Califano
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, M20 4BX, UK; Division of Cancer Sciences, The University of Manchester, Manchester, M13 9NT, UK.
| |
Collapse
|
6
|
Shi H, Guo N, Zhao Z, Liu L, Ni T, Zhang J, Lu Y. Comparison of the second-line treatments for patients with small cell lung cancer sensitive to previous platinum-based chemotherapy: A systematic review and Bayesian network analysis. Front Oncol 2023; 13:1154685. [PMID: 37007093 PMCID: PMC10061131 DOI: 10.3389/fonc.2023.1154685] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 03/06/2023] [Indexed: 03/18/2023] Open
Abstract
ObjectiveIt remains unclear what the best second-line treatment is for patients with small-cell lung cancer sensitive to previous platinum-based chemotherapy.MethodsWe systematically screened randomized controlled trials from several online databases. The primary outcome was objective response rate (ORR), and the secondary outcomes were disease control rate (DCR), overall survival (OS), progression-free survival (PFS), and hematological complications graded 3 to 5. The efficacy of included treatments was ranked by surface under the cumulative ranking curve (SUCRA) value.ResultsWe included eleven trials involving 1560 patients in quantitative analysis. Triple chemotherapy containing platinum (TP, combination of cisplatin, etoposide, and irinotecan) was associated with favorable ORR (intravenous topotecan vs TP; odds ratio: 0.13, 95% CI:0.03-0.63; SUCRA, 0.94) and PFS (vs intravenous topotecan; hazard ratio, 0.5; 95% CI: 0.25-0.99; SUCRA, 0.90). Belotecan ranked highest for OS (SUCRA, 0.90), while intravenous topotecan plus Ziv-aflibercept ranked highest for DCR (SUCRA, 0.75). TP was more likely to cause anemia and thrombocytopenia while intravenous topotecan plus Ziv-aflibercept resulted in most neutrocytopenia.ConclusionTP is the first recommendation for the second-line treatment of sensitive relapsed SCLC. TP achieved priority in ORR and PFS with the most frequent adverse effects in anemia and thrombocytopenia. For patients who cannot tolerate the hematological adverse effects of triple chemotherapy, amrubicin is an optional option. Amrubicin had relatively good ORR and PFS, accompanied by fewer hematological complications. The rechallenge of the platinum doublet is inferior to amrubicin in ORR, DCR, and PFS. Oral topotecan has a similar effect compared with IV topotecan, but oral topotecan was associated with slightly higher safety and less stress in nursing. Belotecan contributed to the best PFS with slightly better safety but was not ideal in other outcomes.Systematic review registrationhttps://www.crd.york.ac.uk/PROSPERO/, identifier CRD42022358256.
Collapse
Affiliation(s)
- Hekai Shi
- Department of Thoracic Surgery, Fudan University Affiliated Huadong Hospital, Shanghai, China
| | - Nuojin Guo
- Shanghai East Hospital, Tongji University, Shanghai, China
| | - Zeming Zhao
- Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Ligang Liu
- Institute of Therapeutic Innovations and Outcomes, College of Pharmacy, The Ohio State University, Columbus, OH, United States
| | - Tianyi Ni
- Department of Thoracic Surgery, Fudan University Affiliated Huadong Hospital, Shanghai, China
| | - Jinye Zhang
- First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yingjie Lu
- Department of Thoracic Surgery, Fudan University Affiliated Huadong Hospital, Shanghai, China
- *Correspondence: Yingjie Lu,
| |
Collapse
|
7
|
Bernabé-Caro R, Chen Y, Dowlati A, Eason P. Current and Emerging Treatment Options for Patients With Relapsed Small-cell Lung Carcinoma: A Systematic Literature Review. Clin Lung Cancer 2023; 24:185-208. [PMID: 36907793 DOI: 10.1016/j.cllc.2023.01.012] [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: 09/29/2022] [Revised: 01/16/2023] [Accepted: 01/20/2023] [Indexed: 02/10/2023]
Abstract
Second-line treatment options are limited for patients with small-cell lung cancer (SCLC). We conducted a PRISMA-standard systematic literature review to evaluate the treatment landscape for patients with relapsed SCLC (PROSPERO number: CRD42022299759). Systematic searches of MEDLINE, Embase, and Cochrane Library were performed (October 2022) to identify publications (prior 5 years) from prospective studies of therapies for relapsed SCLC. Publications were screened against predetermined eligibility criteria; data were extracted to standardized fields. Publication quality was assessed using GRADE. The data were analyzed descriptively, grouped by drug class. Overall, 77 publications involving 6349 patients were included. Studies of tyrosine kinase inhibitors (TKIs) with established cancer indications accounted for 24 publications; topoisomerase I inhibitors for 15; checkpoint inhibitors (CPIs) for 11, and alkylating agents for 9 publications. The remaining 18 publications featured chemotherapies, small-molecule inhibitors, investigational TKIs and monoclonal antibodies, and a cancer vaccine. According to GRADE assessment, 69% of the publications reported low-/very-low-quality evidence; quality limitations included lack of randomization and small sample sizes. Only 6 publications/6 trials reported phase 3 data; 5 publications/2 trials reported phase 2/3 results. Overall, the clinical potential of alkylating agents and CPIs remained unclear; investigations of combination approaches and biomarker-directed usage are warranted. Phase 2 data from TKI trials were consistently promising; no phase 3 data were available. Phase 2 data for a liposomal formulation of irinotecan were promising. We confirmed an absence of promising investigational drug/regimens in late-stage development; thus, relapsed SCLC remains an area of high unmet need.
Collapse
Affiliation(s)
| | - Yuanbin Chen
- Cancer & Hematology Centers of Western Michigan, Grand Rapids, MI
| | - Afshin Dowlati
- Division of Hematology and Oncology, Department of Medicine, Case Western Reserve University, Cleveland, OH; University Hospitals Seidman Cancer Center, Cleveland, OH
| | | |
Collapse
|
8
|
Rosner S, Levy B. Relapsed small-cell lung cancer: a disease of continued unmet need. THE LANCET. RESPIRATORY MEDICINE 2023; 11:6-8. [PMID: 36252600 DOI: 10.1016/s2213-2600(22)00389-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 09/15/2022] [Indexed: 12/24/2022]
Affiliation(s)
- Samuel Rosner
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Benjamin Levy
- Sidney Kimmel Comprehensive Cancer Center at Sibley Memorial Hospital, Washington, DC 20016, USA.
| |
Collapse
|
9
|
Aix SP, Ciuleanu TE, Navarro A, Cousin S, Bonanno L, Smit EF, Chiappori A, Olmedo ME, Horvath I, Grohé C, Farago AF, López-Vilariño JA, Cullell-Young M, Nieto A, Vasco N, Gómez J, Kahatt C, Zeaiter A, Carcereny E, Roubec J, Syrigos K, Lo G, Barneto I, Pope A, Sánchez A, Kattan J, Zarogoulidis K, Waller CF, Bischoff H, Juan-Vidal O, Reinmuth N, Dómine M, Paz-Ares L. Combination lurbinectedin and doxorubicin versus physician's choice of chemotherapy in patients with relapsed small-cell lung cancer (ATLANTIS): a multicentre, randomised, open-label, phase 3 trial. THE LANCET. RESPIRATORY MEDICINE 2023; 11:74-86. [PMID: 36252599 DOI: 10.1016/s2213-2600(22)00309-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 08/16/2022] [Accepted: 08/17/2022] [Indexed: 11/08/2022]
Abstract
BACKGROUND Lurbinectedin is a synthetic marine-derived anticancer agent that acts as a selective inhibitor of oncogenic transcription. Lurbinectedin monotherapy (3·2 mg/m2 every 3 weeks) received accelerated approval from the US Food and Drug Administration on the basis of efficacy in patients with small-cell lung cancer (SCLC) who relapsed after first-line platinum-based chemotherapy. The ATLANTIS trial assessed the efficacy and safety of combination lurbinectedin and the anthracycline doxorubicin as second-line treatment for SCLC. METHODS In this phase 3, open-label, randomised study, adult patients aged 18 years or older with SCLC who relapsed after platinum-based chemotherapy were recruited from 135 hospitals across North America, South America, Europe, and the Middle East. Patients were randomly assigned (1:1) centrally by dynamic allocation to intravenous lurbinectedin 2·0 mg/m2 plus doxorubicin 40·0 mg/m2 administered on day 1 of 21-day cycles or physician's choice of control therapy (intravenous topotecan 1·5 mg/m2 on days 1-5 of 21-day cycles; or intravenous cyclophosphamide 1000 mg/m2, doxorubicin 45·0 mg/m2, and vincristine 2·0 mg on day 1 of 21-day cycles [CAV]) administered until disease progression or unacceptable toxicity. Primary granulocyte-colony stimulating factor prophylaxis was mandatory in both treatment groups. Neither patients nor clinicians were masked to treatment allocation, but the independent review committee, which assessed outcomes, was masked to patients' treatment allocation. The primary endpoint was overall survival in the intention-to-treat population. This trial is registered with ClinicalTrials.gov, NCT02566993, and with EudraCT, 2015-001641-89, and is complete. FINDINGS Between Aug 30, 2016, and Aug 20, 2018, 613 patients were randomly assigned to lurbinectedin plus doxorubicin (n=307) or control (topotecan, n=127; CAV, n=179) and comprised the intention-to-treat population; safety endpoints were assessed in patients who had received any partial or complete study treatment infusions (lurbinectedin plus doxorubicin, n=303; control, n=289). After a median follow-up of 24·1 months (95% CI 21·7-26·3), 303 patients in the lurbinectedin plus doxorubicin group and 289 patients in the control group had discontinued study treatment; progressive disease was the most common reason for discontinuation (213 [70%] patients in the lurbinectedin plus doxorubicin group vs 152 [53%] in the control group). Median overall survival was 8·6 months (95% CI 7·1-9·4) in the lurbinectedin plus doxorubicin group versus 7·6 months (6·6-8·2) in the control group (stratified log-rank p=0·90; hazard ratio 0·97 [95% CI 0·82-1·15], p=0·70). 12 patients died because of treatment-related adverse events: two (<1%) of 303 in the lurbinectedin plus doxorubicin group and ten (3%) of 289 in the control group. 296 (98%) of 303 patients in the lurbinectedin plus doxorubicin group had treatment-emergent adverse events compared with 284 (98%) of 289 patients in the control group; treatment-related adverse events occurred in 268 (88%) patients in the lurbinectedin plus doxorubicin group and 266 (92%) patients in the control group. Grade 3 or worse haematological adverse events were less frequent in the lurbinectedin plus doxorubicin group than the control group (anaemia, 57 [19%] of 302 patients in the lurbinectedin plus doxorubicin group vs 110 [38%] of 288 in the control group; neutropenia, 112 [37%] vs 200 [69%]; thrombocytopenia, 42 [14%] vs 90 [31%]). The frequency of treatment-related adverse events leading to treatment discontinuation was lower in the lurbinectedin plus doxorubicin group than in the control group (26 [9%] of 303 patients in the lurbinectedin plus doxorubicin group vs 47 [16%] of 289 in the control group). INTERPRETATION Combination therapy with lurbinectedin plus doxorubicin did not improve overall survival versus control in patients with relapsed SCLC. However, lurbinectedin plus doxorubicin showed a favourable haematological safety profile compared with control. FUNDING PharmaMar.
Collapse
Affiliation(s)
- Santiago Ponce Aix
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Tudor Eliade Ciuleanu
- Department of Oncology, Institutul Oncologic Prof Dr Ion Chiricuta, Cluj-Napoca, Romania
| | - Alejandro Navarro
- Medical Oncology Department, Vall d'Hebron University Hospital & Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Sophie Cousin
- Department of Medical Oncology, Institut Bergonié, Bordeaux, France
| | - Laura Bonanno
- Medical Oncology 2, Istituto Oncologico Veneto, Padova, Italy
| | - Egbert F Smit
- Department of Thoracic Oncology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Alberto Chiappori
- Department of Thoracic Oncology, H Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | | | - Ildiko Horvath
- National Koranyi Institute for Pulmonology, Budapest, Hungary
| | - Christian Grohé
- Department of Respiratory Diseases, Evangelische Lungenklinik Berlin, Berlin, Germany
| | - Anna F Farago
- Harvard Medical School, Boston, MA, USA; Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | | | | | | | | | | | | | | | - Enric Carcereny
- Badalona-Applied Research Group in Oncology (B-ARGO) and Medical Oncology Department, Catalan Institute of Oncology Badalona, Germans Trias i Pujol Hospital, Barcelona, Spain
| | - Jaromir Roubec
- Nemocnice AGEL Ostrava-Vítkovice, Ostrava-Vítkovice, Czech Republic
| | | | - Gregory Lo
- R S McLaughlin Durham Regional Cancer Centre, Lakeridge Health, Oshawa, ON, Canada
| | | | - Anthony Pope
- Clatterbridge Cancer Centre NHS Foundation Trust, Wirral, UK
| | | | - Joseph Kattan
- Hotel-Dieu de France University Hospital, Saint Joseph University, Beirut, Lebanon
| | | | | | - Helge Bischoff
- Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany
| | - Oscar Juan-Vidal
- Department of Medical Oncology, Hospital Universitario La Fe, Valencia, Spain
| | | | - Manuel Dómine
- Hospital Universitario Fundación Jiménez Diaz, Madrid, Spain
| | - Luis Paz-Ares
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain; CNIO-H12o Lung Cancer Clinical Research Unit, Madrid, Spain; Ciberonc, Madrid, Spain; Universidad Complutense de Madrid, Madrid, Spain.
| |
Collapse
|
10
|
Challenges in the treatment of small cell lung cancer in the era of immunotherapy and molecular classification. Lung Cancer 2023; 175:88-100. [PMID: 36493578 DOI: 10.1016/j.lungcan.2022.11.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/18/2022] [Accepted: 11/20/2022] [Indexed: 11/26/2022]
Abstract
For many years the standard of care for small cell lung cancer (SCLC) has remained unchanged. Despite decades of active research, current treatment options are limited and the prognosis of patients with extended disease (ED) SCLC remains poor. The introduction of immune checkpoint inhibitors (ICIs) represents an exception and the only recent approval for ED-SCLC. However, the magnitude of benefit obtained with immunotherapy in SCLC is much more modest than that observed in other malignancies. Different pro-immunogenic or immunosuppressive features within the tumor microenvironment of SCLC may either modulate the sensitivity to immunotherapy or conversely dampen the efficacy of ICIs. Beside immunotherapy, a deeper understanding of the molecular biology of SCLC has led to the identification of new therapeutic targets for this lethal malignancy. Recent epigenetic and gene expression studies have resulted into a new molecular classification of four distinct subtypes of SCLC, defined by the relative expression of key transcription regulators and each characterized by specific therapeutic vulnerabilities. This review discusses the rationale for immunotherapy in SCLC and summarizes the main ICIs-trials in this tumor. We provide also an overview of new potential therapeutic opportunities and their integration with the new molecular classification of SCLC.
Collapse
|
11
|
Rittberg R, Leung B, Al-Hashami Z, Ho C. Real World Patient Eligibility for Second Line Lurbinectedin Based Treatment in Small Cell Lung Cancer: Understanding Epidemiology and Estimating Health Care Utilization. Curr Oncol 2022; 29:9744-9752. [PMID: 36547179 PMCID: PMC9776936 DOI: 10.3390/curroncol29120765] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/25/2022] [Accepted: 12/02/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND In the ATLANTIS study, second-line lurbinectedin/doxorubicin did not improve overall survival (OS), however patients with a chemotherapy-free interval (CTFI) of ≥180 days had an improved progression free survival (PFS). The objective of this retrospective study was to identify the proportion of real-world small cell lung cancer (SCLC) patients who are suitable for lurbinectedin-based therapy based on these criteria. METHODS A retrospective study of all SCLC referred to BC Cancer between 2012 and 2017 was conducted. Patient demographics, staging, treatment, and survival data were collected retrospectively. Baseline characteristics were compared using descriptive statistics. OS was calculated using Kaplan-Meier curves. Statistically significant p-value was <0.05. RESULTS A total of 1048 patients were identified. Baseline characteristics: median age 68 years, 47% male, 61% current smoking status, 68% extensive disease. Best supportive care was received by 22%. First-line systemic therapy was platinum doublet for 71% of the population. Second-line systemic therapy was delivered to 22%. Of the 219 patients who received second-line systemic therapy after platinum doublet, 183 patients had a CTFI of ≥90 days and 107 patients had a CTFI of ≥180 days. Patients originally treated as limited stage disease, received platinum doublet as second line, received thoracic radiation (RT) or prophylactic cranial irradiation (PCI) were more likely to have a CTFI of ≥90 and ≥180 days. CONCLUSION In our real-world SCLC population, only 21% of the SCLC population received second-line therapy after platinum doublet with 17% achieving CTFI of ≥90 days and 10% CTFI of ≥180 days. Based on this retrospective review, only a small fraction of platinum-treated patients would be preferentially offered lurbinectedin in the second-line setting.
Collapse
Affiliation(s)
- Rebekah Rittberg
- BC Cancer, Department of Medical Oncology, Vancouver, BC V5Z 4E6, Canada
- CancerCare Manitoba, Section of Hematology/Oncology, Department of Internal Medicine, University of Manitoba, Winnipeg, MB R3E 0V9, Canada
| | - Bonnie Leung
- BC Cancer, Department of Medical Oncology, Vancouver, BC V5Z 4E6, Canada
| | - Zamzam Al-Hashami
- Sultan Qaboos Comprehensive Cancer Care and Research Center, Muscat P.O. Box 566 P.C 123, Oman
| | - Cheryl Ho
- BC Cancer, Department of Medical Oncology, Vancouver, BC V5Z 4E6, Canada
| |
Collapse
|
12
|
Wang N, Yu C, Xu T, Yao D, Zhu L, Shen Z, Huang X. Self-assembly of DNA nanostructure containing cell-specific aptamer as a precise drug delivery system for cancer therapy in non-small cell lung cancer. J Nanobiotechnology 2022; 20:486. [PMID: 36403038 PMCID: PMC9675138 DOI: 10.1186/s12951-022-01701-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 11/09/2022] [Indexed: 11/21/2022] Open
Abstract
Background As the most common subtype in lung cancer, the precise and efficient treatment for non-small cell lung cancer (NSCLC) remains an outstanding challenge owing to early metastasis and poor prognosis. Chemotherapy, the most commonly used treatment modality, is a difficult choice for many cancer patients due to insufficient drug accumulation in tumor sites and severe systemic side-effects. In this study, we constructed a cell-specific aptamer-modified DNA nanostructure (Apt-NS) as a targeting drug delivery system achieving the precision therapy for lung cancer. Methods The synthesis of DNA nanostructure and its stability were evaluated using gel electrophoresis. The targeting properties and internalization mechanism were investigated via flow cytometry and confocal analyses. Drug loading, release, and targeted drug delivery were determined by fluorescence detection, Zeta potentials assay, and confocal imaging. CCK8 assays, colony formation, cell apoptosis, metastasis analyses and in vivo experiments were conducted to assess the biological functions of DNA nanostructure. Results Self-assembled DNA nanoparticles (Apt-NS) had excellent stability to serum and DNase I and the ability to specifically recognize A549 cells. Upon specific binding, the drug-loaded nanoparticles (Apt-NS-DOX) were internalized into target cells by clathrin-mediated endocytosis. Subsequently, DOX could be released from Apt-NS-DOX based on the degradation of the lysosome. Apt-NS-DOX exerted significant suppression of cell proliferation, invasion and migration, and also enhanced cell apoptosis due to the excellent performance of drug delivery and intracellular release, while maintaining a superior biosafety. In addition, the antitumor effects of Apt-NS-DOX were further confirmed using in vivo models. Conclusions Our study provided cell-specific aptamer-modified DNA nanostructures as a drug-delivery system targeting A549 cells, which could precisely and efficiently transport chemotherapeutic drug into tumor cells, exerting enhanced antineoplastic efficacy. These findings highlight that DNA nanostructure serving as an ideal drug delivery system in cancer treatment appears great promise in biomedical applications. Supplementary Information The online version contains supplementary material available at 10.1186/s12951-022-01701-5.
Collapse
Affiliation(s)
- Ning Wang
- grid.414906.e0000 0004 1808 0918Division of Pulmonary Medicine, the First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325000 China
| | - Chang Yu
- grid.414906.e0000 0004 1808 0918Intervention Department, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000 China
| | - Tingting Xu
- grid.414906.e0000 0004 1808 0918Division of Pulmonary Medicine, the First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325000 China
| | - Dan Yao
- grid.414906.e0000 0004 1808 0918Division of Pulmonary Medicine, the First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325000 China
| | - Lingye Zhu
- grid.414906.e0000 0004 1808 0918Division of Pulmonary Medicine, the First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325000 China
| | - Zhifa Shen
- grid.268099.c0000 0001 0348 3990Key Laboratory of Laboratory Medicine, Ministry of Education of China, and Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035 China
| | - Xiaoying Huang
- grid.414906.e0000 0004 1808 0918Division of Pulmonary Medicine, the First Affiliated Hospital of Wenzhou Medical University, Key Laboratory of Heart and Lung, Wenzhou, Zhejiang 325000 China
| |
Collapse
|
13
|
Gadducci A, Cosio S. Trabectedin and lurbinectedin: Mechanisms of action, clinical impact, and future perspectives in uterine and soft tissue sarcoma, ovarian carcinoma, and endometrial carcinoma. Front Oncol 2022; 12:914342. [PMID: 36408147 PMCID: PMC9671549 DOI: 10.3389/fonc.2022.914342] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 10/12/2022] [Indexed: 11/06/2022] Open
Abstract
The ecteinascidins trabectedin and lurbinectedin are very interesting antineoplastic agents, with a favorable toxicity profile and peculiar mechanisms of action. These drugs form adducts in the minor groove of DNA, which produce single-strand breaks (SSBs) and double-strand breaks (DSBs) and trigger a series of events resulting in cell cycle arrest and apoptosis. Moreover, the ecteinascidins interact with the tumor microenvironment, reduce the number of tumor-associated macrophages, and inhibit the secretion of cytokines and chemokines. Trabectedin has been approved by the Federal Drug Administration (FDA) for patients with unresectable or metastatic liposarcoma or leiomyosarcoma who received a prior anthracycline-based regimen. Moreover, trabectedin in combination with pegylated liposomal doxorubicin (PLD) has been approved in the European Union for the treatment of platinum-sensitive recurrent ovarian cancer. Lurbinectedin has been approved by the FDA for patients with metastatic small cell lung cancer with disease progression on or after platinum-based chemotherapy. The review assesses in vitro and in vivo experimental studies on the antineoplastic effects of both ecteinascidins as well as the clinical trials on the activity of trabectedin in uterine sarcoma and ovarian carcinoma and of lurbinectedin in ovarian carcinoma and endometrial carcinoma.
Collapse
|
14
|
Manzo A, Sforza V, Carillio G, Palumbo G, Montanino A, Sandomenico C, Costanzo R, Esposito G, Laudato F, Mercadante E, La Manna C, Muto P, Totaro G, De Cecio R, Picone C, Piccirillo MC, Pascarella G, Normanno N, Morabito A. Lurbinectedin in small cell lung cancer. Front Oncol 2022; 12:932105. [PMID: 36110944 PMCID: PMC9469650 DOI: 10.3389/fonc.2022.932105] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 08/08/2022] [Indexed: 11/25/2022] Open
Abstract
Few treatment options are available for patients with small cell lung cancer (SCLC) in progression after a first-line therapy. A novel therapeutic approach is represented by lurbinectedin, a synthetic derivative of trabectedin that works by inhibiting oncogenic transcription and promoting apoptosis in tumor cells. A phase II basket trial demonstrated the activity of lurbinectedin at the dose of 3.2 mg/m2 in patients with SCLC who had failed a previous chemotherapy, with a response rate of 35.2%, a median progression-free survival (mPFS) of 3.5 months, and a median overall survival (mOS) of 9.3 months. Common severe adverse events (grades 3-4) were hematological disorders, including anemia (9%), leukopenia (29%), neutropenia (46%), and thrombocytopenia (7%). On the basis of the positive results of this phase II study, on June 2020, lurbinectedin was approved by the Food and Drug Administration as second line for SCLC patients in progression on or after platinum-based therapy. The subsequent phase III trial comparing the combination of lurbinectedin plus doxorubicin vs. CAV (cyclophosphamide, Adriamycin, and vincristine) or topotecan did not demonstrate an improvement in overall survival, although the experimental arm showed a superior safety profile. Combinations of lurbinectedin with other drugs, cytotoxic agents and immune checkpoint inhibitors, are currently under investigation. The results of these studies should better define the optimal clinical application of lurbinectedin.
Collapse
Affiliation(s)
- Anna Manzo
- Thoracic Medical Oncology, Istituto Nazionale Tumori, “Fondazione G. Pascale” - IRCCS, Napoli, Italy
| | - Vincenzo Sforza
- Thoracic Medical Oncology, Istituto Nazionale Tumori, “Fondazione G. Pascale” - IRCCS, Napoli, Italy
| | - Guido Carillio
- Department of Oncology and Hematology, Azienda Ospedaliera Pugliese-Ciaccio, Catanzaro, Italy
| | - Giuliano Palumbo
- Thoracic Medical Oncology, Istituto Nazionale Tumori, “Fondazione G. Pascale” - IRCCS, Napoli, Italy
| | - Agnese Montanino
- Thoracic Medical Oncology, Istituto Nazionale Tumori, “Fondazione G. Pascale” - IRCCS, Napoli, Italy
| | - Claudia Sandomenico
- Thoracic Medical Oncology, Istituto Nazionale Tumori, “Fondazione G. Pascale” - IRCCS, Napoli, Italy
| | - Raffaele Costanzo
- Thoracic Medical Oncology, Istituto Nazionale Tumori, “Fondazione G. Pascale” - IRCCS, Napoli, Italy
| | | | - Francesca Laudato
- Thoracic Medical Oncology, Istituto Nazionale Tumori, “Fondazione G. Pascale” - IRCCS, Napoli, Italy
| | - Edoardo Mercadante
- Thoracic Surgery, Istituto Nazionale Tumori, “Fondazione G. Pascale” – IRCCS, Napoli, Italy
| | - Carmine La Manna
- Thoracic Surgery, Istituto Nazionale Tumori, “Fondazione G. Pascale” – IRCCS, Napoli, Italy
| | - Paolo Muto
- Radiotherapy, Istituto Nazionale Tumori “Fondazione G. Pascale” - IRCCS, Naples –, Italy
| | - Giuseppe Totaro
- Radiotherapy, Istituto Nazionale Tumori “Fondazione G. Pascale” - IRCCS, Naples –, Italy
| | - Rossella De Cecio
- Pathology, Istituto Nazionale Tumori, “Fondazione G. Pascale” – IRCCS, Napoli, Italy
| | - Carmine Picone
- Radiology, Istituto Nazionale Tumori, “Fondazione G. Pascale” – IRCCS, Napoli, Italy
| | | | - Giacomo Pascarella
- Scientific Directorate, Istituto Nazionale Tumori “Fondazione G. Pascale” - IRCCS, Napoli, Italy
| | - Nicola Normanno
- Scientific Directorate, Istituto Nazionale Tumori “Fondazione G. Pascale” - IRCCS, Napoli, Italy
- Cellular Biology and Biotherapy, Istituto Nazionale Tumori, “Fondazione G. Pascale” – IRCCS, Napoli, Italy
| | - Alessandro Morabito
- Thoracic Medical Oncology, Istituto Nazionale Tumori, “Fondazione G. Pascale” - IRCCS, Napoli, Italy
| |
Collapse
|
15
|
Pangua C, Rogado J, Serrano-Montero G, Belda-Sanchís J, Álvarez Rodríguez B, Torrado L, Rodríguez De Dios N, Mielgo-Rubio X, Trujillo JC, Couñago F. New perspectives in the management of small cell lung cancer. World J Clin Oncol 2022; 13:429-447. [PMID: 35949427 PMCID: PMC9244973 DOI: 10.5306/wjco.v13.i6.429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 09/05/2021] [Accepted: 05/22/2022] [Indexed: 02/06/2023] Open
Abstract
The treatment of small cell lung cancer (SCLC) is a challenge for all specialists involved. New treatments have been added to the therapeutic armamentarium in recent months, but efforts must continue to improve both survival and quality of life. Advances in surgery and radiotherapy have resulted in prolonged survival times and fewer complications, while more careful patient selection has led to increased staging accuracy. Developments in the field of systemic therapy have resulted in changes to clinical guidelines and the management of patients with advanced disease, mainly with the introduction of immunotherapy. In this article, we describe recent improvements in the management of patients with SCLC, review current treatments, and discuss future lines of research.
Collapse
Affiliation(s)
- Cristina Pangua
- Department of Medical Oncology, Hospital Universitario Infanta Leonor, Madrid 28031, Spain
| | - Jacobo Rogado
- Department of Medical Oncology, Hospital Universitario Infanta Leonor, Madrid 28031, Spain
| | - Gloria Serrano-Montero
- Department of Medical Oncology, Hospital Universitario Infanta Leonor, Madrid 28031, Spain
| | - José Belda-Sanchís
- Department of Thoracic Surgery, Hospital de la Santa Creu i Sant Pau & Hospital de Mar, Universitat Autònoma de Barcelona, Barcelona 08041, Catalonia, Spain
| | - Beatriz Álvarez Rodríguez
- Department of Radiation Oncology, Hospital Universitario HM Sanchinarro, HM Hospitales, HM CIOCC Centro Integral Oncológico Clara Campal, Madrid 28050, Spain
| | - Laura Torrado
- Department of Radiation Oncology, Hospital Universitario Lucus Augusti & Instituto de Investigación Sanitaria Santiago de Compostela (IDIS), Lugo 27003, Spain
| | - Nuria Rodríguez De Dios
- Department of Radiation Oncology, Hospital Del Mar & Hospital Del Mar Medical Research Institute (IMIM) & Pompeu Fabra University, Barcelona 08003, Catalonia, Spain
| | - Xabier Mielgo-Rubio
- Department of Medical Oncology, Alcorcón Foundation University Hospital, Alcorcón 28922, Madrid, Spain
| | - Juan Carlos Trujillo
- Department of Thoracic Surgery, Hospital de la Santa Creu i Sant Pau, Barcelona 08029, Spain
| | - Felipe Couñago
- Department of Radiation Oncology, Hospital Universitario Quirónsalud Madrid, Hospital La Luz, Universidad Europea de Madrid, Madrid 28223, Spain
| |
Collapse
|
16
|
Kulkarni NS, Gupta V. Repurposing therapeutics for malignant pleural mesothelioma (MPM) - Updates on clinical translations and future outlook. Life Sci 2022; 304:120716. [PMID: 35709894 DOI: 10.1016/j.lfs.2022.120716] [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: 04/15/2022] [Revised: 05/31/2022] [Accepted: 06/09/2022] [Indexed: 10/18/2022]
Abstract
INTRODUCTION Malignant pleural mesothelioma (MPM) is a rare malignancy affecting the mesothelial cells in the pleural lining surrounding the lungs. First approved chemotherapy against MPM was a platinum/antifolate (cisplatin/pemetrexed) (2003). Since then, no USFDA approvals have gone through for small molecules as these molecules have not been proven to be therapeutically able in later stages of clinical studies. An alternative to conventional chemotherapy can be utilization of monoclonal antibodies, which are proven to improve patient survival significantly as compared to conventional chemotherapy (Nivolumab + Ipilimumab, 2020). AREA COVERED Drug repurposing has been instrumental in drug discovery for rare diseases such as MPM and multiple repositioned small molecule therapies and immunotherapies are currently being tested for its applicability in MPM management. This article summarizes essential breakthroughs along the pre-clinical and clinical developmental stages of small molecules and monoclonal antibodies for MPM management. EXPERT OPINION For rare diseases such as malignant pleural mesothelioma, a drug repurposing strategy can be adapted as it eases the financial burden on pharmaceutical companies along with fast-tracking development. With the rise of multiple small molecule repurposed therapies and innovations in localized treatment, MPM therapeutics are bound to be more effective in this decade.
Collapse
Affiliation(s)
- Nishant S Kulkarni
- College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Vivek Gupta
- College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA.
| |
Collapse
|
17
|
Randomized Phase 3 Study of the Anti-disialoganglioside Antibody Dinutuximab and Irinotecan vs Irinotecan or Topotecan for Second-Line Treatment of Small Cell Lung Cancer. Lung Cancer 2022; 166:135-142. [DOI: 10.1016/j.lungcan.2022.03.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 02/24/2022] [Accepted: 03/02/2022] [Indexed: 01/22/2023]
|
18
|
Zugazagoitia J, Paz-Ares L. Extensive-Stage Small-Cell Lung Cancer: First-Line and Second-Line Treatment Options. J Clin Oncol 2022; 40:671-680. [PMID: 34985925 DOI: 10.1200/jco.21.01881] [Citation(s) in RCA: 81] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Extensive-stage small-cell lung cancer is a therapeutically challenging disease. After more than two decades without clinical progress, the addition of programmed cell death protein 1 axis blockade to platinum-based chemotherapy has demonstrated sustained overall survival benefit and represents the current standard of care in the first-line setting. Despite this benefit, resistance emerges relatively rapidly in virtually all patients. Although newer treatments are being incorporated in the relapse setting, marked therapeutic resistance is typically observed in patients with relapsed small-cell lung cancer (SCLC), underscoring the need of developing more effective therapies in this setting. Notably, recent progress in the understanding of the molecular biology of SCLC might bring possibilities toward molecularly informed therapeutic strategies for patients with SCLC, which could have a significant impact for improving outcomes in this disease. Here, we review current treatment options and recent progress made in the first-line and relapsed SCLC, including the role of biomarkers and new evolving therapeutic strategies.
Collapse
Affiliation(s)
- Jon Zugazagoitia
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain.,H12O-CNIO Lung Cancer Clinical Research Unit, Health Research Institute Hospital Universitario 12 de Octubre (i+12)/Spanish National Cancer Research Center (CNIO), Madrid, Spain.,CIBERONC, Madrid, Spain
| | - Luis Paz-Ares
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain.,H12O-CNIO Lung Cancer Clinical Research Unit, Health Research Institute Hospital Universitario 12 de Octubre (i+12)/Spanish National Cancer Research Center (CNIO), Madrid, Spain.,CIBERONC, Madrid, Spain.,Department of Medicine, Complutense University, Madrid, Spain
| |
Collapse
|
19
|
Tan AC, Boggs DH, Lee EQ, Kim MM, Mehta MP, Khasraw M. Clinical Trial Eligibility Criteria and Recently Approved Cancer Therapies for Patients With Brain Metastases. Front Oncol 2022; 11:780379. [PMID: 35047397 PMCID: PMC8761732 DOI: 10.3389/fonc.2021.780379] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 12/09/2021] [Indexed: 12/24/2022] Open
Abstract
Brain metastases cause significant morbidity and mortality in patients with advanced cancer. In the era of precision oncology and immunotherapy, there are rapidly evolving systemic treatment options. These novel therapies may have variable intracranial efficacy, and patients with brain metastases remain a population of special interest. Typically, only patients with stable, asymptomatic and/or treated brain metastases are enrolled in clinical trials, or may be excluded altogether, particularly in the setting of leptomeningeal carcinomatosis. Consequently, this leads to significant concerns on the external validity of clinical trial evidence to real-world clinical practice. Here we describe the current trends in cancer clinical trial eligibility for patients with brain metastases in both early and late phase trials, with a focus on targeted and immunotherapies. We evaluate recent newly FDA approved therapies and the clinical trial evidence base leading to approval. This includes analysis of inclusion and exclusion criteria, requirements for baseline screening for brain metastases, surveillance cerebral imaging and incorporation of trial endpoints for patients with brain metastases. Finally, the use of alternative sources of data such as real-world evidence with registries and collaborative studies will be discussed.
Collapse
Affiliation(s)
- Aaron C Tan
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore.,Duke-NUS Medical School, National University of Singapore, Singapore, Singapore
| | - Drexell H Boggs
- Department of Radiation Oncology, University of Alabama at Birmingham School of Medicine, Birmingham, AL, United States
| | - Eudocia Q Lee
- Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Michelle M Kim
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, United States
| | - Minesh P Mehta
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, FL, United States
| | - Mustafa Khasraw
- Duke Cancer Institute, Duke University, Durham, NC, United States
| |
Collapse
|
20
|
Binnewies M, Pollack JL, Rudolph J, Dash S, Abushawish M, Lee T, Jahchan NS, Canaday P, Lu E, Norng M, Mankikar S, Liu VM, Du X, Chen A, Mehta R, Palmer R, Juric V, Liang L, Baker KP, Reyno L, Krummel MF, Streuli M, Sriram V. Targeting TREM2 on tumor-associated macrophages enhances immunotherapy. Cell Rep 2021; 37:109844. [PMID: 34686340 DOI: 10.1016/j.celrep.2021.109844] [Citation(s) in RCA: 133] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 05/09/2021] [Accepted: 09/27/2021] [Indexed: 01/22/2023] Open
Abstract
Converting checkpoint inhibitor (CPI)-resistant individuals to being responsive requires identifying suppressive mechanisms. We identify TREM2+ tumor-associated macrophages (TAMs) as being correlated with exhausted CD8+ tumor-infiltrating lymphocytes (TILs) in mouse syngeneic tumor models and human solid tumors of multiple histological types. Fc domain-enhanced anti-TREM2 monoclonal antibody (mAb) therapy promotes anti-tumor immunity by elimination and modulation of TAM populations, which leads to enhanced CD8+ TIL infiltration and effector function. TREM2+ TAMs are most enriched in individuals with ovarian cancer, where TREM2 expression corresponds to disease grade accompanied by worse recurrence-free survival. In an aggressive orthotopic ovarian cancer model, anti-TREM2 mAb therapy drives potent anti-tumor immunity. These results highlight TREM2 as a highly attractive target for immunotherapy modulation in individuals who are refractory to CPI therapy and likely have a TAM-rich tumor microenvironment.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents, Immunological/pharmacology
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- CD8-Positive T-Lymphocytes/drug effects
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Cell Line, Tumor
- Coculture Techniques
- Drug Resistance, Neoplasm
- Female
- HEK293 Cells
- Humans
- Immune Checkpoint Inhibitors/pharmacology
- Lymphocyte Activation/drug effects
- Lymphocytes, Tumor-Infiltrating/drug effects
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Membrane Glycoproteins/antagonists & inhibitors
- Membrane Glycoproteins/metabolism
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Neoplasms/drug therapy
- Neoplasms/immunology
- Neoplasms/metabolism
- Neoplasms/pathology
- Programmed Cell Death 1 Receptor/antagonists & inhibitors
- Programmed Cell Death 1 Receptor/immunology
- Programmed Cell Death 1 Receptor/metabolism
- Receptors, Immunologic/antagonists & inhibitors
- Receptors, Immunologic/metabolism
- Signal Transduction
- Tumor Cells, Cultured
- Tumor Microenvironment
- Tumor-Associated Macrophages/drug effects
- Tumor-Associated Macrophages/immunology
- Tumor-Associated Macrophages/metabolism
- Mice
Collapse
Affiliation(s)
| | | | - Joshua Rudolph
- Pionyr Immunotherapeutics, South San Francisco, CA 94080, USA
| | - Subhadra Dash
- Pionyr Immunotherapeutics, South San Francisco, CA 94080, USA
| | | | - Tian Lee
- Pionyr Immunotherapeutics, South San Francisco, CA 94080, USA
| | | | - Pamela Canaday
- Pionyr Immunotherapeutics, South San Francisco, CA 94080, USA
| | - Erick Lu
- Pionyr Immunotherapeutics, South San Francisco, CA 94080, USA
| | - Manith Norng
- Pionyr Immunotherapeutics, South San Francisco, CA 94080, USA
| | - Shilpa Mankikar
- Pionyr Immunotherapeutics, South San Francisco, CA 94080, USA
| | - Victoria M Liu
- Pionyr Immunotherapeutics, South San Francisco, CA 94080, USA
| | - Xiaoyan Du
- Pionyr Immunotherapeutics, South San Francisco, CA 94080, USA
| | - Amanda Chen
- Pionyr Immunotherapeutics, South San Francisco, CA 94080, USA
| | - Ranna Mehta
- Pionyr Immunotherapeutics, South San Francisco, CA 94080, USA
| | - Rachael Palmer
- Pionyr Immunotherapeutics, South San Francisco, CA 94080, USA
| | | | - Linda Liang
- Pionyr Immunotherapeutics, South San Francisco, CA 94080, USA
| | - Kevin P Baker
- Pionyr Immunotherapeutics, South San Francisco, CA 94080, USA.
| | - Leonard Reyno
- Pionyr Immunotherapeutics, South San Francisco, CA 94080, USA
| | - Matthew F Krummel
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA.
| | - Michel Streuli
- Pionyr Immunotherapeutics, South San Francisco, CA 94080, USA
| | | |
Collapse
|
21
|
Kristeleit R, Moreno V, Boni V, Guerra EM, Kahatt C, Romero I, Calvo E, Basté N, López-Vilariño JA, Siguero M, Alfaro V, Zeaiter A, Forster M. Doxorubicin plus lurbinectedin in patients with advanced endometrial cancer: results from an expanded phase I study. Int J Gynecol Cancer 2021; 31:1428-1436. [PMID: 34610971 PMCID: PMC8573419 DOI: 10.1136/ijgc-2021-002881] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 09/03/2021] [Indexed: 11/10/2022] Open
Abstract
Objective Second-line treatment of endometrial cancer is an unmet medical need. We conducted a phase I study evaluating lurbinectedin and doxorubicin intravenously every 3 weeks in patients with solid tumors. The aim of this study was to characterise the efficacy and safety of lurbinectedin and doxorubicin for patients with endometrial cancer. Methods Thirty-four patients were treated: 15 patients in the escalation phase (doxorubicin 50 mg/m2 and lurbinectedin 3.0–5.0 mg) and 19 patients in the expansion cohort (doxorubicin 40 mg/m2 and lurbinectedin 2.0 mg/m2). All histological subtypes were eligible and patients had received one to two prior lines of chemotherapy for advanced disease. Antitumor activity was evaluated every two cycles according to the Response Evaluation Criteria in Solid Tumors version 1.1. Adverse events were graded according to the National Cancer Institute-Common Terminology Criteria for Adverse Events version 4. Results Median age (range) was 65 (51–78) years. Eastern Cooperative Oncology Group performance status was up to 1 in 97% of patients. In the escalation phase, 4 (26.7%) of 15 patients had confirmed response: two complete and two partial responses (95% CI 7.8% to 55.1%). Median duration of response was 19.5 months. Median progression-free survival was 7.3 (2.5 to 10.1) months. In the expansion cohort, confirmed partial response was reported in 8 (42.1%) of 19 patients (95% CI 20.3% to 66.5%). Median duration of response was 7.5 (6.4 to not reached) months, median progression-free survival was 7.7 (2.0 to 16.7) months and median overall survival was 14.2 (4.5 to not reached) months. Fatigue (26.3% of patients), and transient and reversible myelosuppression (neutropenia, 78.9%; febrile neutropenia, 21.1%; thrombocytopenia, 15.8%) were the main grade 3 and higher toxicities in the expanded cohort. Conclusions In patients with recurrent advanced endometrial cancer treated with doxorubicin and lurbinectedin, response rates (42%) and duration of response (7.5 months) were favorable. Further evaluation of doxorubicin and lurbinectedin is warranted in this patient population.
Collapse
Affiliation(s)
| | - Victor Moreno
- START Madrid-FJD, Hospital Fundación Jiménez Díaz, Madrid, Spain
| | - Valentina Boni
- START Madrid-CIOCC, Centro Integral Oncológico Clara Campal, Madrid, Spain
| | - Eva M Guerra
- Hospital Universitario Ramón y Cajal, Madrid, Spain
| | | | | | - Emiliano Calvo
- START Madrid-CIOCC, Centro Integral Oncológico Clara Campal, Madrid, Spain
| | - Neus Basté
- Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | | | | | - Vicente Alfaro
- Clinical Development, PharmaMar SA, Colmenar Viejo, Spain
| | | | | |
Collapse
|
22
|
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: 0.7] [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
|
23
|
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: 78] [Impact Index Per Article: 26.0] [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
|
24
|
Zhou X, Guan Z, Jin X, Zhao J, Chen G, Ding J, Ren Y, Zhai X, Zhou Q, Guan Z. Reversal of alopecia areata, osteoporosis follow treatment with activation of Tgr5 in mice. Biosci Rep 2021; 41:BSR20210609. [PMID: 34196345 PMCID: PMC8292761 DOI: 10.1042/bsr20210609] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 04/28/2021] [Accepted: 04/30/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Alopecia areata is an autoimmune hair loss disease with infiltration of pro-inflammatory cells into hair follicles. The role of Tgr5 in dermatitis has attracted considerable attention. The present study aimed to investigate the effect of Tgr5 in the development of Alopecia areata. METHODS The study utilized a comparison control group design with four groups of wild-type group, wild-type+INT777 group, Tgr5-/- group, and Tgr5-/-+INT777 group. The mice were treated with INT777 (30 mg/kg/day) or the carrier solution (DMSO) intraperitoneally for 7 weeks, and the back skin was collected and analyzed by histology and immunohistochemistry staining. The lumbar vertebrae 4 has also been analyzed by DXA and Micro-CT. RESULTS Tgr5-/- mice displayed the decreasingly significant in hair area and length, skin thickness, and the ratio of anagen and telogen, collagen, and mast cell number and loss the bone mass than WT group. After treating with INT777, the appearance of alopecia areata and bone microstructure has improved. Immunohistochemistry and qPCR analysis showed that activation of Tgr5 can down-regulate the express of JAK1, STAT3, IL-6, TNF-α, and VEGF. CONCLUSION These findings indicate that activation of Tgr5 mediated amelioration of alopecia areata and osteoporosis by down-regulated JAK1-STAT3 signaling pathway.
Collapse
Affiliation(s)
- Xiaohui Zhou
- Qinghai Provincial People’s Hospital, Xining, Qinghai, P.R. China
| | - Zhiqiang Guan
- Department of Dermatology, The First People's Hospital of Xuzhou, Xuzhou, Jiangsu 221002, P.R. China
| | - Xiao Jin
- Department of Rheumatology and Immunology, The First People’s Hospital of Xuzhou, Xuzhou, Jiangsu 221002, P.R. China
| | - Jianbin Zhao
- Department of Dermatology, The First People's Hospital of Xuzhou, Xuzhou, Jiangsu 221002, P.R. China
| | - Guisheng Chen
- Department of Dermatology, Traditional Chinese Medicine Hospital of Xuzhou Jiangsu 221002, P.R. China
| | - Jicun Ding
- Department of Dermatology, The First People's Hospital of Xuzhou, Xuzhou, Jiangsu 221002, P.R. China
| | - Yile Ren
- Department of Rheumatology and Immunology, The First People’s Hospital of Xuzhou, Xuzhou, Jiangsu 221002, P.R. China
| | - Xiaoxiang Zhai
- Department of Dermatology, The Seventh People’s Hospital of Shanghai, Shanghai 200137, P.R. China
| | - Qiyun Zhou
- Qinghai Provincial People’s Hospital, Xining, Qinghai, P.R. China
| | - Zhiyuan Guan
- Department of Orthopedics, The Shanghai Tenth People's Hospital of Tongji University, Shanghai, P.R. China
- Department of Orthopedics, Peking University Third Hospital, Beijing, P.R. China
| |
Collapse
|
25
|
Metastatic small cell lung cancer - an aggressive disease: a case report and literature review. Anticancer Drugs 2021; 32:1138-1141. [PMID: 34232947 DOI: 10.1097/cad.0000000000001119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
In the present case study, we describe a 53-year-old male with an aggressive small cell lung cancer (SCLC) that was diagnosed in January 2019. Our patient was treated as first line of systemic chemotherapy consisting of cisplatin and etoposide followed by mediastinal prophylactic radiotherapy with good response later he received for his metastatic disease (M-SCLC) a rechallenge of systemic chemotherapy consisting of carboplatin, etoposide and dulvalumab with stable disease and after progression his disease he was treated with lurbinectedin and after four cycles he reached a complete radiologic response. To the best of our knowledge, this is the first case to be reported of M-SCLC patient treated with prior of two types of platinum combination with immunotherapy and reaching a complete radiologic response.
Collapse
|
26
|
Hou K, Shen J, Yan J, Zhai C, Zhang J, Pan JA, Zhang Y, Jiang Y, Wang Y, Lin RZ, Cong H, Gao S, Zong WX. Loss of TRIM21 alleviates cardiotoxicity by suppressing ferroptosis induced by the chemotherapeutic agent doxorubicin. EBioMedicine 2021; 69:103456. [PMID: 34233258 PMCID: PMC8261003 DOI: 10.1016/j.ebiom.2021.103456] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 06/09/2021] [Accepted: 06/10/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Doxorubicin, an anthracycline chemotherapeutic agent, is widely used in the treatment of many cancers. However, doxorubicin posts a great risk of adverse cardiovascular events, which are thought to be caused by oxidative stress. We recently reported that the ubiquitin E3 ligase TRIM21 interacts and ubiquitylates p62 and negatively regulates the p62-Keap1-Nrf2 antioxidant pathway. Therefore, we sought to determine the role TRIM21 in cardiotoxicity induced by oxidative damage. METHODS Using TRIM21 knockout mice, we examined the effects of TRIM21 on cardiotoxicity induced by two oxidative damage models: the doxorubicin treatment model and the Left Anterior Descending (LAD) model. We also explored the underlying mechanism by RNA-sequencing of the heart tissues, and by treating the mouse embryonic fibroblasts (MEFs), immortalized rat cardiomyocyte line H9c2, and immortalized human cardiomyocyte line AC16 with doxorubicin. FINDINGS TRIM21 knockout mice are protected from heart failure and fatality in both the doxorubicin and LAD models. Hearts of doxorubicin-treated wild-type mice exhibit deformed mitochondria and elevated level of lipid peroxidation reminiscent of ferroptosis, which is alleviated in TRIM21 knockout hearts. Mechanistically, TRIM21-deficient heart tissues and cultured MEFs and H9c2 cells display enhanced p62 sequestration of Keap1 and are protected from doxorubicin-induced ferroptosis. Reconstitution of wild-type but not the E3 ligase-dead and the p62 binding-deficient TRIM21 mutants impedes the protection from doxorubicin-induced cell death. INTERPRETATION Our study demonstrates that TRIM21 ablation protects doxorubicin-induced cardiotoxicity and illustrates a new function of TRIM21 in ferroptosis, and suggests TRIM21 as a therapeutic target for reducing chemotherapy-related cardiotoxicity. FUNDING NIH (CA129536; DK108989): data collection, analysis. Shanghai Pujiang Program (19PJ1401900): data collection. National Natural Science Foundation (31971161): data collection. Department of Veteran Affairs (BX004083): data collection. Tianjin Science and Technology Plan Project (17ZXMFSY00020): data collection.
Collapse
Affiliation(s)
- Kai Hou
- School of Medicine, Nankai University, Tianjin, China; Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, China; Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, USA; Department of Cardiology, Tianjin Chest Hospital, Tianjin, China
| | - Jianliang Shen
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, USA
| | - Junrong Yan
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, USA
| | - Chuannan Zhai
- School of Medicine, Nankai University, Tianjin, China; Department of Cardiology, Tianjin Chest Hospital, Tianjin, China
| | - Jingxia Zhang
- Department of Cardiology, Tianjin Chest Hospital, Tianjin, China
| | - Ji-An Pan
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, Sun Yat-sen University, Shenzhen, China
| | - Ye Zhang
- Tianjin Third Central Hospital, Tianjin, China
| | - Yaping Jiang
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY, USA
| | - Yongbo Wang
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Richard Z Lin
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY, USA
| | - Hongliang Cong
- School of Medicine, Nankai University, Tianjin, China; Department of Cardiology, Tianjin Chest Hospital, Tianjin, China.
| | - Shenglan Gao
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, China.
| | - Wei-Xing Zong
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, USA; Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA.
| |
Collapse
|
27
|
Wahab A, Rafae A, Mushtaq K, Venkata K, Sarmad R. Lurbinectedin-Induced Tumor Lysis Syndrome in Small Cell Neuroendocrine Cancer of the Cecum: A First-Ever Case Report. AMERICAN JOURNAL OF CASE REPORTS 2021; 22:e932081. [PMID: 34125741 PMCID: PMC8212840 DOI: 10.12659/ajcr.932081] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Patient: Female, 38-year-old Final Diagnosis: Tumor lysis syndrome Symptoms: Abdominal pain Medication:— Clinical Procedure: — Specialty: Hematology • Oncology
Collapse
Affiliation(s)
- Ahsan Wahab
- Department of Hospital Medicine/Internal Medicine, Baptist Medical Center South, Montgomery, AL, USA
| | - Abdul Rafae
- Department of Internal Medicine, McLaren Regional Medical Center, Flint, MI, USA
| | - Kamran Mushtaq
- Department of Hospital Medicine, Northeast Internal Medicine Associates, LaGrange, IN, USA
| | - Krishna Venkata
- UAB Montgomery Internal Medicine Residency, Baptist Medical Center South, Montgomery, AL, USA
| | - Rehan Sarmad
- Department of Hematology-Oncology, Alabama Oncology Hematology Associates, Montgomery, AL, USA
| |
Collapse
|
28
|
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: 1.0] [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
|
29
|
Patel S, Petty WJ, Sands JM. An overview of lurbinectedin as a new second-line treatment option for small cell lung cancer. Ther Adv Med Oncol 2021; 13:17588359211020529. [PMID: 34104228 PMCID: PMC8165873 DOI: 10.1177/17588359211020529] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 05/05/2021] [Indexed: 12/16/2022] Open
Abstract
Small cell lung cancer (SCLC) is a highly proliferative, aggressive form of lung cancer that carries a poor prognosis. Recent approvals with new therapeutic options represent the first in more than a decade for SCLC. Lurbinectedin, a newly approved second-line option, is a synthetic alkaloid that covalently binds DNA, generating double-strand breaks, and disrupts DNA-protein interactions and RNA transcription. Lurbinectedin may also modulate the tumor microenvironment by inducing apoptosis of peripheral blood monocytes and tumor associated macrophages, decreasing expression of the inflammatory chemokine (C-C motif) ligand 2 (CCL2) and reducing tumor angiogenesis. A single-arm, open-label, basket trial included 105 patients with SCLC that had received one prior line of therapy. Patients received lurbinectedin 3.2 mg/m2 as an intravenous infusion every 3 weeks, resulting in a response rate of 35.2% and a disease control rate of 68.6%. The response rate was 45% among those with >90 days chemotherapy free interval (CTFI) and 22% in the resistant group (CTFI < 90 days). The median overall survival was 9.3 months. Myelosuppression is the most frequent clinically significant adverse event, particularly neutropenia; however, neutropenic fever occurred in only 5% of those in the SCLC cohort of the basket trial. Nausea and fatigue were also noted. The side effect profile compares favorably to topotecan, while a direct comparison of tolerability can be made between lurbinectedin versus topotecan or pegylated-liposomal doxorubicin from CORAIL, a randomized study for platinum-resistant/refractory ovarian cancer. A press release has reported the ongoing clinical trial for SCLC including combination lurbinectedin and doxorubicin versus topotecan or cyclophosphamide, doxorubicin, and vinblastine to be negative. The details may provide more insight at publication, and future trials will be important to further define the clinical utility of lurbinectedin. Lurbinectedin represents a new option in second-line SCLC.
Collapse
Affiliation(s)
- Shetal Patel
- Department of Medicine, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - William Jeffrey Petty
- Hematology and Oncology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Jacob M Sands
- Thoracic Oncology, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA 02215, USA Department of Medicine, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
30
|
Bocharova EA, Kopytina NI, Slynko ЕЕ. Anti-tumour drugs of marine origin currently at various stages of clinical trials (review). REGULATORY MECHANISMS IN BIOSYSTEMS 2021. [DOI: 10.15421/022136] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Oncological diseases for a long time have remained one of the most significant health problems of modern society, which causes great losses in its labour and vital potential. Contemporary oncology still faces unsolved issues as insufficient efficacy of treatment of progressing and metastatic cancer, chemoresistance, and side-effects of the traditional therapy which lead to disabilities among or death of a high number of patients. Development of new anti-tumour preparations with a broad range of pharmaceutical properties and low toxicity is becoming increasingly relevant every year. The objective of the study was to provide a review of the recent data about anti-tumour preparations of marine origin currently being at various phases of clinical trials in order to present the biological value of marine organisms – producers of cytotoxic compounds, and the perspectives of their use in modern biomedical technologies. Unlike the synthetic oncological preparations, natural compounds are safer, have broader range of cytotoxic activity, can inhibit the processes of tumour development and metastasis, and at the same time have effects on several etiopathogenic links of carcinogenesis. Currently, practical oncology uses 12 anti-tumour preparations of marine origin (Fludarabine, Cytarabine, Midostaurin, Nelarabine, Eribulin mesylate, Brentuximab vedotin, Trabectedin, Plitidepsin, Enfortumab vedotin, Polatuzumab vedotin, Belantamab mafodotin, Lurbinectedin), 27 substances are at different stages of clinical trials. Contemporary approaches to the treatment of oncological diseases are based on targeted methods such as immune and genetic therapies, antibody-drug conjugates, nanoparticles of biopolymers, and metals. All those methods employ bioactive compounds of marine origin. Numerous literature data from recent years indicate heightened attention to the marine pharmacology and the high potential of marine organisms for the biomedicinal and pharmaceutic industries.
Collapse
|
31
|
Evaluation of the Preclinical Efficacy of Lurbinectedin in Malignant Pleural Mesothelioma. Cancers (Basel) 2021; 13:cancers13102332. [PMID: 34066159 PMCID: PMC8151304 DOI: 10.3390/cancers13102332] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/09/2021] [Accepted: 05/10/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary The marine drug lurbinectedin revealed an unprecedented efficacy against patient-derived malignant pleural mesothelioma cells, regardless of the histological type and the BAP1 mutation status. By inducing strong DNA damages, it dramatically arrested cell cycle progression and induced apoptosis. These results may be translated into the use of lurbinectedin as an effective agent for malignant pleural mesothelioma patients. Abstract Background: Malignant pleural mesothelioma (MPM) is a highly aggressive cancer generally diagnosed at an advanced stage and characterized by a poor prognosis. The absence of alterations in druggable kinases, together with an immune-suppressive tumor microenvironment, limits the use of molecular targeted therapies, making the treatment of MPM particularly challenging. Here we investigated the in vitro susceptibility of MPM to lurbinectedin (PM01183), a marine-derived drug that recently received accelerated approval by the FDA for the treatment of patients with metastatic small cell lung cancer with disease progression on or after platinum-based chemotherapy. Methods: A panel of primary MPM cultures, resembling the three major MPM histological subtypes (epithelioid, sarcomatoid, and biphasic), was characterized in terms of BAP1 status and histological markers. Subsequently, we explored the effects of lurbinectedin at nanomolar concentration on cell cycle, cell viability, DNA damage, genotoxic stress response, and proliferation. Results: Stabilized MPM cultures exhibited high sensitivity to lurbinectedin independently from the BAP1 mutational status and histological classification. Specifically, we observed that lurbinectedin rapidly promoted a cell cycle arrest in the S-phase and the activation of the DNA damage response, two conditions that invariably resulted in an irreversible DNA fragmentation, together with strong apoptotic cell death. Moreover, the analysis of long-term treatment indicated that lurbinectedin severely impacts MPM transforming abilities in vitro. Conclusion: Overall, our data provide evidence that lurbinectedin exerts a potent antitumoral activity on primary MPM cells, independently from both the histological subtype and BAP1 alteration, suggesting its potential activity in the treatment of MPM patients.
Collapse
|
32
|
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: 12] [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
|
33
|
Olmedo ME, Forster M, Moreno V, López-Criado MP, Braña I, Flynn M, Doger B, de Miguel M, López-Vilariño JA, Núñez R, Kahatt C, Cullell-Young M, Zeaiter A, Calvo E. Efficacy and safety of lurbinectedin and doxorubicin in relapsed small cell lung cancer. Results from an expansion cohort of a phase I study. Invest New Drugs 2021; 39:1275-1283. [PMID: 33704620 PMCID: PMC8426303 DOI: 10.1007/s10637-020-01025-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 10/22/2020] [Indexed: 11/25/2022]
Abstract
Background A phase I study found remarkable activity and manageable toxicity for doxorubicin (bolus) plus lurbinectedin (1-h intravenous [i.v.] infusion) on Day 1 every three weeks (q3wk) as second-line therapy in relapsed small cell lung cancer (SCLC). An expansion cohort further evaluated this combination. Patients and methods Twenty-eight patients with relapsed SCLC after no more than one line of cytotoxic-containing chemotherapy were treated: 18 (64%) with sensitive disease (chemotherapy-free interval [CTFI] ≥90 days) and ten (36%) with resistant disease (CTFI <90 days; including six with refractory disease [CTFI ≤30 days]). Results Ten patients showed confirmed response (overall response rate [ORR] = 36%); median progression-free survival (PFS) = 3.3 months; median overall survival (OS) = 7.9 months. ORR was 50% in sensitive disease (median PFS = 5.7 months; median OS = 11.5 months) and 10% in resistant disease (median PFS = 1.3 months; median OS = 4.6 months). The main toxicity was transient and reversible myelosuppression. Treatment-related non-hematological events (fatigue, nausea, decreased appetite, vomiting, alopecia) were mostly mild or moderate. Conclusion Doxorubicin 40 mg/m2 and lurbinectedin 2.0 mg/m2 on Day 1 q3wk has shown noteworthy activity in relapsed SCLC and a manageable safety profile. The combination is being evaluated as second-line therapy for SCLC in an ongoing, randomized phase III trial. Clinical trial registration www.ClinicalTrials.gov code: NCT01970540. Date of registration: 22 October, 2013.
Collapse
Affiliation(s)
| | - Martin Forster
- University College of London Hospital and UCL Cancer Institute, London, UK
| | - Victor Moreno
- START Madrid - FJD (Hospital Fundación Jiménez Díaz), Madrid, Spain
| | | | - Irene Braña
- Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Michael Flynn
- University College of London Hospital and UCL Cancer Institute, London, UK
| | - Bernard Doger
- START Madrid - FJD (Hospital Fundación Jiménez Díaz), Madrid, Spain
| | - María de Miguel
- START Madrid - HM CIOCC, Hospital Madrid Norte Sanchinarro, Madrid, Spain
| | | | | | | | | | - Ali Zeaiter
- Pharma Mar, S.A., Colmenar Viejo, Madrid, Spain
| | - Emiliano Calvo
- START Madrid - HM CIOCC, Hospital Madrid Norte Sanchinarro, Madrid, Spain.
| |
Collapse
|
34
|
Cortinovis D, Bidoli P, Canova S, Colonese F, Gemelli M, Lavitrano ML, Banna GL, Liu SV, Morabito A. Novel Cytotoxic Chemotherapies in Small Cell Lung Carcinoma. Cancers (Basel) 2021; 13:1152. [PMID: 33800236 PMCID: PMC7962524 DOI: 10.3390/cancers13051152] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 02/24/2021] [Accepted: 03/03/2021] [Indexed: 01/08/2023] Open
Abstract
Small cell lung cancer (SCLC) is one of the deadliest thoracic neoplasms, in part due to its fast doubling time and early metastatic spread. Historically, cytotoxic chemotherapy consisting of platinum-etoposide or anthracycline-based regimens has demonstrated a high response rate, but early chemoresistance leads to a poor prognosis in advanced SCLC. Only a fraction of patients with limited-disease can be cured by chemo-radiotherapy. Given the disappointing survival rates in advanced SCLC, new cytotoxic agents are eagerly awaited. Unfortunately, few novel chemotherapy drugs have been developed in the latest decades. This review describes the results and potential application in the clinical practice of novel chemotherapy agents for SCLC.
Collapse
Affiliation(s)
- Diego Cortinovis
- Department Medical Oncology—ASST-Monza Ospedale San Gerardo, via Pergolesi 33, 20090 Monza, Italy; (P.B.); (S.C.); (F.C.); (M.G.)
| | - Paolo Bidoli
- Department Medical Oncology—ASST-Monza Ospedale San Gerardo, via Pergolesi 33, 20090 Monza, Italy; (P.B.); (S.C.); (F.C.); (M.G.)
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy;
| | - Stefania Canova
- Department Medical Oncology—ASST-Monza Ospedale San Gerardo, via Pergolesi 33, 20090 Monza, Italy; (P.B.); (S.C.); (F.C.); (M.G.)
| | - Francesca Colonese
- Department Medical Oncology—ASST-Monza Ospedale San Gerardo, via Pergolesi 33, 20090 Monza, Italy; (P.B.); (S.C.); (F.C.); (M.G.)
| | - Maria Gemelli
- Department Medical Oncology—ASST-Monza Ospedale San Gerardo, via Pergolesi 33, 20090 Monza, Italy; (P.B.); (S.C.); (F.C.); (M.G.)
| | | | - Giuseppe Luigi Banna
- Department of Oncology, Portsmouth Hospitals University NHS Trust, Cosham, Portsmouth PO6 3LY, UK;
| | - Stephen V. Liu
- Lombardi Comprehensive Cancer Center, Georgetown University, 3800 Reservoir Road NW, Washington, DC 20007, USA;
| | - Alessandro Morabito
- SC Oncologia Medica Toraco-Polmonare, IRCCS Istituto Nazionale dei Tumori, Fondazione Pascale, 80100 Napoli, Italy;
| |
Collapse
|
35
|
Pierret T, Toffart AC, Giaj Levra M, Moro-Sibilot D, Gobbini E. Advances and Therapeutic Perspectives in Extended-Stage Small-Cell Lung Cancer. Cancers (Basel) 2020; 12:E3224. [PMID: 33139612 PMCID: PMC7692868 DOI: 10.3390/cancers12113224] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/27/2020] [Accepted: 10/30/2020] [Indexed: 01/13/2023] Open
Abstract
Extended small cell lung cancer (ED-SCLC) is a very aggressive disease, characterized by rapid growth and an early tendency to relapse. In contrast to non-small cell lung cancer, no therapeutic innovation has improved survival in patients with ED-SCLC over the past 20 years. Recently, immunotherapy has shown an important role in the management of these patients, emerging as the treatment of first choice in combination with chemotherapy and completely changing the therapeutic paradigm. However, patients' selection for this strategy is still challenging due to a lack of reliable predictive biomarkers. Conversely, the immunotherapy efficacy beyond the first line is pretty disappointing and innovative chemotherapies or target agents seem to be more promising in this setting. Some of them are also under evaluation as an upfront strategy and they will probably change the treatment algorithm in the next future. This proposal provides a comprehensive overview of available treatment strategies for ED-SCLC patients, highlighting their strengths and weaknesses.
Collapse
Affiliation(s)
- Thomas Pierret
- Thoracic Oncology Unit, CHU Grenoble-Alpes, 38700 Grenoble, France; (T.P.); (A.-C.T.); (M.G.L.); (D.M.-S.)
| | - Anne-Claire Toffart
- Thoracic Oncology Unit, CHU Grenoble-Alpes, 38700 Grenoble, France; (T.P.); (A.-C.T.); (M.G.L.); (D.M.-S.)
| | - Matteo Giaj Levra
- Thoracic Oncology Unit, CHU Grenoble-Alpes, 38700 Grenoble, France; (T.P.); (A.-C.T.); (M.G.L.); (D.M.-S.)
| | - Denis Moro-Sibilot
- Thoracic Oncology Unit, CHU Grenoble-Alpes, 38700 Grenoble, France; (T.P.); (A.-C.T.); (M.G.L.); (D.M.-S.)
| | - Elisa Gobbini
- Thoracic Oncology Unit, CHU Grenoble-Alpes, 38700 Grenoble, France; (T.P.); (A.-C.T.); (M.G.L.); (D.M.-S.)
- Cancer Research Center of Lyon, 69008 Lyon, France
| |
Collapse
|
36
|
Abstract
Small-cell lung cancer has defied our scientific community for decades. Chemotherapy has been the mainstay treatment for small-cell lung cancer (SCLC) and unlike its counterpart, non-small cell lung cancer, no significant therapeutic breakthroughs have been made since the 1970s. Among the reasons for this slow-paced therapeutic development, one that stands out is the distinctive and almost universal loss of function of the tumour suppressor genes TP53 and RB1 in this disease, for which pharmacological activation has yet to be achieved, despite having been highly sought after. Although no molecularly targeted approach has been approved for clinical practice thus far, several strategies are currently exploring the potential to drug the tumour's "Achilles heel" that stems from essential pathways regulating DNA-damage response. Most recently, we have witnessed newfound reasons to hope, as the combination of immunotherapy and systemic chemotherapy has improved survival outcomes, representing the first landmark achievement in decades and a new standard of care for patients with extensive disease SCLC. However, continuous efforts are still needed towards a better understanding of the molecular pathways that singularise this tumour to eventually identify the predictive biomarkers that might result in the development of a more rational therapeutic approach, including the use of immunotherapy combinations. In this review we aim to uncover critical aspects of the immune microenvironment and biology of SCLC and provide an overview of the current and future landscape of promising therapeutic opportunities. The challenge still stands, but regardless, we are living in exciting times to finally check SCLC off the "bucket list" of our scientific community.
Collapse
|
37
|
Serzan MT, Farid S, Liu SV. Drugs in development for small cell lung cancer. J Thorac Dis 2020; 12:6298-6307. [PMID: 33209468 PMCID: PMC7656445 DOI: 10.21037/jtd-2019-sclc-10] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 05/13/2020] [Indexed: 12/30/2022]
Abstract
Small cell lung cancer (SCLC) is a particularly lethal subtype of lung cancer whose treatment landscape has been relatively devoid of advance. The recent integration of immunotherapy in the first-line treatment of SCLC has improved overall survival (OS), prompting the first major paradigm shift for this disease in decades. Despite this improvement in outcomes, most patients with SCLC will relapse after initial response. Standard salvage systemic therapy for SCLC remains disappointing, with few approved agents and consistently poor outcomes. The need for novel agents to combat this disease remains pressing. Fortunately, there are several agents in various stages of development that hold potential as novel treatments for advanced SCLC. Lurbinectedin, which targets active transcription, has shown activity in platinum-sensitive and platinum-resistant SCLC as monotherapy and in combination with doxorubicin. Aurora A kinase (AAK) inhibitors showed initial activity when given with paclitaxel but in randomized studies, failed to improve outcomes over paclitaxel plus placebo. However, in the subset of patients with MYC expression, targeting AAK was effective. Similarly, agents targeting poly-ADP ribose (PARP) pair well with other DNA damaging drugs but in the subset of patients whose tumors express Schlafen-11 (SLFN-11), efficacy appeared greater. CDK 4/6 inhibition is being explored, primarily as a means to protect myeloid cells during cytotoxic chemotherapy in a strategy expected to be uniquely effective in SCLC. Ongoing trials are also studying are novel formulations of established cytotoxic agents. Delta-like protein 3 (DLL3) is an appealing therapeutic target given its selective expression on SCLC cells, but after initial exciting results, the antibody-drug conjugate (ADC) Rovalpituzumab tesirine (Rova-T) did not have a favorable efficacy to toxicity profile in randomized trials. Other agents targeting DLL3 are under study. Targeting angiogenesis has yielded modest improvements in the past but newer agents such as anlotinib are renewing interest. While the current therapeutic landscape beyond chemo-immunotherapy remains the same as it was decades ago, drug development for SCLC is rapidly moving forward and promises to deliver the needed novel agents in the very near future.
Collapse
Affiliation(s)
- Michael T. Serzan
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Saira Farid
- Department of Internal Medicine, Medstar Washington Hospital Center, Washington DC, USA
| | - Stephen V. Liu
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| |
Collapse
|
38
|
Yang M, Li L, Chen S, Li S, Wang B, Zhang C, Chen Y, Yang L, Xin H, Chen C, Xu X, Zhang Q, He Y, Ye J. Melatonin protects against apoptosis of megakaryocytic cells via its receptors and the AKT/mitochondrial/caspase pathway. Aging (Albany NY) 2020; 12:13633-13646. [PMID: 32651992 PMCID: PMC7377846 DOI: 10.18632/aging.103483] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 05/27/2020] [Indexed: 04/19/2023]
Abstract
Clinical studies have shown that melatonin lowers the frequency of thrombocytopenia in patients with cancer undergoing radiotherapy or chemotherapy. Here, we investigated the mechanisms by which melatonin promotes platelet formation and survival. Our results show that melatonin exerted protective effects on serum-free induced apoptosis of CHRF megakaryocytes (MKs). Melatonin promoted the formation of MK colony forming units (CFUs) in a dose-dependent manner. Using doxorubicin-treated CHRF cells, we found that melatonin rescued G2/M cell cycle arrest and cell apoptosis induced by doxorubicin. The expression of p-AKT was increased by melatonin treatment, an effect that was abolished by melatonin receptor blocker. In addition, we demonstrated that melatonin enhanced the recovery of platelets in an irradiated mouse model. Megakaryopoiesis was largely preserved in melatonin-treated mice. We obtained the same results in vivo from bone marrow histology and CFU-MK formation assays. Melatonin may exert these protective effects by directly stimulating megakaryopoiesis and inhibiting megakaryocyte apoptosis through activation of its receptors and AKT signaling.
Collapse
Affiliation(s)
- Mo Yang
- The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, China
- Nanfang Hospital, Southern Medical University, Guangzhou, China
- Lianjiang People’s Hospital, Lianjiang, Guangdong, China
| | - Liang Li
- The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Shichao Chen
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Suyi Li
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Bo Wang
- The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Changhua Zhang
- The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, China
- The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Youpeng Chen
- The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Liuming Yang
- Lianjiang People’s Hospital, Lianjiang, Guangdong, China
| | - Hongwu Xin
- Lianjiang People’s Hospital, Lianjiang, Guangdong, China
| | - Chun Chen
- The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Xiaojun Xu
- The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Qing Zhang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Yulong He
- The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Jieyu Ye
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
39
|
Portugal J. Insights into DNA-drug interactions in the era of omics. Biopolymers 2020; 112:e23385. [PMID: 32542701 DOI: 10.1002/bip.23385] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 05/23/2020] [Accepted: 05/25/2020] [Indexed: 01/07/2023]
Abstract
Despite the rise of sophisticated new targeting strategies in cancer chemotherapy, many classic DNA-binding drugs remain on the front line of the therapy against cancer. Based on examples primarily from the author's laboratory, this article reviews the capabilities of several DNA-binding drugs to alter gene expression. Research is ongoing about the molecular bases of the inhibition of gene expression and how alteration of the cellular transcriptome can commit cancer cells to die. The development of a variety of omic techniques allows us to gain insights into the effect of antitumor drugs. Genome-wide approaches provide unbiased genomic data that can facilitate a deeper understanding of the cellular response to DNA-binding drugs. Moreover, the results of large-scale genomic studies are gathered in publicly available databases that can be used in developing precision medicine in cancer treatment.
Collapse
Affiliation(s)
- José Portugal
- Instituto de Diagnóstico Ambiental y Estudios del Agua, CSIC, Barcelona, Spain
| |
Collapse
|
40
|
Chen P, Kuang P, Wang L, Li W, Chen B, Liu Y, Wang H, Zhao S, Ye L, Yu F, He Y, Zhou C. Mechanisms of drugs-resistance in small cell lung cancer: DNA-related, RNA-related, apoptosis-related, drug accumulation and metabolism procedure. Transl Lung Cancer Res 2020; 9:768-786. [PMID: 32676338 PMCID: PMC7354133 DOI: 10.21037/tlcr-19-547] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Small-cell lung cancer (SCLC), the highest malignant cancer amongst different types of lung cancer, has the feature of lower differentiation, rapid growth, and poor survival rate. Despite the dramatically initial sensitivity of SCLC to various types of treatment methods, including chemotherapy, radiotherapy and immunotherapy, the emergence of drugs-resistance is still a grandly clinical challenge. Therefore, in order to improve the prognosis and develop new therapeutic approaches, having a better understanding of the complex mechanisms of resistance in SCLC is of great clinical significance. This review summarized recent advances in understanding of multiple mechanisms which are involved in the resistance during SCLC treatment, including DNA-related process, RNA-related process, apoptosis-related mechanism, and the process of drug accumulation and metabolism.
Collapse
Affiliation(s)
- Peixin Chen
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China.,Department of Medical School, Tongji University, Shanghai, China
| | - Peng Kuang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China.,Department of Medical Oncology, The First Affiliated Hospital Of Nanchang University, Nanchang, China
| | - Lei Wang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Wei Li
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Bin Chen
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Yu Liu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China.,Department of Medical School, Tongji University, Shanghai, China
| | - Hao Wang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China.,Department of Medical School, Tongji University, Shanghai, China
| | - Sha Zhao
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Lingyun Ye
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Feng Yu
- Department of Medical Oncology, The First Affiliated Hospital Of Nanchang University, Nanchang, China
| | - Yayi He
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Caicun Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
41
|
Fernandez-Teruel C, Gonzalez I, Trocóniz IF, Lubomirov R, Soto A, Fudio S. Population-Pharmacokinetic and Covariate Analysis of Lurbinectedin (PM01183), a New RNA Polymerase II Inhibitor, in Pooled Phase I/II Trials in Patients with Cancer. Clin Pharmacokinet 2020; 58:363-374. [PMID: 30090974 DOI: 10.1007/s40262-018-0701-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND OBJECTIVES Lurbinectedin is an inhibitor of RNA polymerase II currently under clinical development for intravenous administration as a single agent and in combination with other anti-tumor agents for the treatment of several tumor types. The objective of this work was to develop a population-pharmacokinetic model in this patient setting and to elucidate the main predictors to guide the late stages of development. METHODS Data from 443 patients with solid and hematologic malignancies treated in six phase I and three phase II trials with lurbinectedin as a single agent or combined with other agents were included in the analysis. The potential influence of demographic, co-treatment, and laboratory characteristics on lurbinectedin pharmacokinetics was evaluated. RESULTS The final population-pharmacokinetic model was an open three-compartment model with linear distribution and linear elimination from the central compartment. Population estimates for total plasma clearance, and apparent volume at steady state were 11.2 L/h and 438 L, respectively. Inter-individual variability was moderate for all parameters, ranging from 20.9 to 51.2%. High α-1-acid glycoprotein and C-reactive protein, and low albumin reduced clearance by 28, 20, and 20%, respectively. Co-administration of cytochrome P450 3A inhibitors reduced clearance by 30%. Combinations with other anti-tumor agents did not modify the pharmacokinetics of lurbinectedin significantly. CONCLUSION The population-pharmacokinetic model indicated neither a dose nor time dependency, and no clinically meaningful pharmacokinetic differences were found when co-administered with other anticancer agents. A chronic inflammation pattern characterized by decreased albumin and increased C-reactive protein and α-1-acid glycoprotein levels led to high lurbinectedin exposure. Co-administration of cytochrome P450 3A inhibitors increased lurbinectedin exposure.
Collapse
Affiliation(s)
- Carlos Fernandez-Teruel
- Department of Clinical Pharmacology, PharmaMar, S.A., Avda. de los Reyes, 1 Colmenar Viejo, 28770, Madrid, Spain.
| | - Ignacio Gonzalez
- Department of Clinical Pharmacology, PharmaMar, S.A., Avda. de los Reyes, 1 Colmenar Viejo, 28770, Madrid, Spain
| | - Iñaki F Trocóniz
- Pharmacometrics and Systems Pharmacology, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain.,IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Rubin Lubomirov
- Department of Clinical Pharmacology, PharmaMar, S.A., Avda. de los Reyes, 1 Colmenar Viejo, 28770, Madrid, Spain
| | - Arturo Soto
- Department of Clinical Pharmacology, PharmaMar, S.A., Avda. de los Reyes, 1 Colmenar Viejo, 28770, Madrid, Spain
| | - Salvador Fudio
- Department of Clinical Pharmacology, PharmaMar, S.A., Avda. de los Reyes, 1 Colmenar Viejo, 28770, Madrid, Spain
| |
Collapse
|
42
|
Taniguchi H, Sen T, Rudin CM. Targeted Therapies and Biomarkers in Small Cell Lung Cancer. Front Oncol 2020; 10:741. [PMID: 32509576 PMCID: PMC7251180 DOI: 10.3389/fonc.2020.00741] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 04/20/2020] [Indexed: 12/13/2022] Open
Abstract
Small cell lung cancer (SCLC) is an aggressive malignancy characterized by rapid growth, early metastasis, and acquired therapeutic resistance. A majority of patients with SCLC have extensive-stage (ES) disease, defined as the presence of metastatic disease outside the hemithorax at first diagnosis. SCLC has been considered “a graveyard for drug development,” with chemotherapy remaining the standard treatment for first- and second-line management until quite recently. In contrast to NSCLC, identifying therapeutic targets in SCLC has been challenging, partly because driver mutations are primarily loss of function, involving the tumor suppressor genes RB1 and TP53 or currently untargetable (e.g., amplification of MYC family members). Recent gene expression profiling of SCLC cells lines, patient samples and representative murine models, have led to a proposed delineation of four major subtypes for SCLC distinguished by differential expression of four key transcriptional regulators (ASCL1, NEUROD1, POU2F3, and YAP1). Our understanding of the biology of SCLC has indeed significantly improved recently due to the continued efforts of the dedicated investigators in this field, but the therapeutic options remain dismal. While recent results from immunotherapy trials are encouraging, most patients demonstrate either primary or rapid acquired resistance to current regimens, highlighting the clear need to improve the effectiveness and expand the scope of current therapeutic strategies. In this opinion article, we will discuss recent developments in the treatment of SCLC, focused on current understanding of the signaling pathways, the role of immunotherapy and targeted therapy, and emerging biomarkers of response to therapy in SCLC.
Collapse
Affiliation(s)
- Hirokazu Taniguchi
- Molecular Pharmacology Program and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States.,Department of Respiratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Triparna Sen
- Molecular Pharmacology Program and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States.,Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| | - Charles M Rudin
- Molecular Pharmacology Program and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States.,Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| |
Collapse
|
43
|
Trigo J, Subbiah V, Besse B, Moreno V, López R, Sala MA, Peters S, Ponce S, Fernández C, Alfaro V, Gómez J, Kahatt C, Zeaiter A, Zaman K, Boni V, Arrondeau J, Martínez M, Delord JP, Awada A, Kristeleit R, Olmedo ME, Wannesson L, Valdivia J, Rubio MJ, Anton A, Sarantopoulos J, Chawla SP, Mosquera-Martinez J, D'Arcangelo M, Santoro A, Villalobos VM, Sands J, Paz-Ares L. Lurbinectedin as second-line treatment for patients with small-cell lung cancer: a single-arm, open-label, phase 2 basket trial. Lancet Oncol 2020; 21:645-654. [DOI: 10.1016/s1470-2045(20)30068-1] [Citation(s) in RCA: 143] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 01/20/2020] [Accepted: 01/22/2020] [Indexed: 12/12/2022]
|
44
|
Abstract
Small cell lung cancer (SCLC) is an aggressive malignancy and carries a poor prognosis with limited effective treatments in the advanced setting. SCLC is characterized by a high tumor mutation burden and alterations in Notch signaling and DNA damage repair pathways, providing rationale for the use of immunotherapy and targeted therapies. Immunotherapies have led to the most significant advances in treating SCLC in decades, and several promising targeted approaches have emerged from the increased understanding of the biology of SCLC. However, responses to these novel approaches are far from universal, and efforts to refine these therapies are ongoing.
Collapse
|
45
|
Poirier JT, George J, Owonikoko TK, Berns A, Brambilla E, Byers LA, Carbone D, Chen HJ, Christensen CL, Dive C, Farago AF, Govindan R, Hann C, Hellmann MD, Horn L, Johnson JE, Ju YS, Kang S, Krasnow M, Lee J, Lee SH, Lehman J, Lok B, Lovly C, MacPherson D, McFadden D, Minna J, Oser M, Park K, Park KS, Pommier Y, Quaranta V, Ready N, Sage J, Scagliotti G, Sos ML, Sutherland KD, Travis WD, Vakoc CR, Wait SJ, Wistuba I, Wong KK, Zhang H, Daigneault J, Wiens J, Rudin CM, Oliver TG. New Approaches to SCLC Therapy: From the Laboratory to the Clinic. J Thorac Oncol 2020; 15:520-540. [PMID: 32018053 PMCID: PMC7263769 DOI: 10.1016/j.jtho.2020.01.016] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 01/10/2020] [Accepted: 01/11/2020] [Indexed: 12/12/2022]
Abstract
The outcomes of patients with SCLC have not yet been substantially impacted by the revolution in precision oncology, primarily owing to a paucity of genetic alterations in actionable driver oncogenes. Nevertheless, systemic therapies that include immunotherapy are beginning to show promise in the clinic. Although, these results are encouraging, many patients do not respond to, or rapidly recur after, current regimens, necessitating alternative or complementary therapeutic strategies. In this review, we discuss ongoing investigations into the pathobiology of this recalcitrant cancer and the therapeutic vulnerabilities that are exposed by the disease state. Included within this discussion, is a snapshot of the current biomarker and clinical trial landscapes for SCLC. Finally, we identify key knowledge gaps that should be addressed to advance the field in pursuit of reduced SCLC mortality. This review largely summarizes work presented at the Third Biennial International Association for the Study of Lung Cancer SCLC Meeting.
Collapse
Affiliation(s)
- John T Poirier
- Perlmutter Cancer Center, New York University Langone Health, New York, New York
| | - Julie George
- Department of Translational Genomics, Center of Integrated Oncology Cologne-Bonn, Medical Faculty, University of Cologne, Cologne Germany
| | | | - Anton Berns
- The Netherlands Cancer Institute, Amsterdam, Netherlands
| | | | | | | | | | | | - Caroline Dive
- Cancer Research United Kingdom, Manchester Institute, Manchester, United Kingdom
| | - Anna F Farago
- Massachusetts General Hospital, Boston, Massachusetts
| | | | - Christine Hann
- Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Leora Horn
- Vanderbilt University, Nashville, Tennessee
| | | | | | - Sumin Kang
- Emory University, Winship Cancer Institute, Atlanta, Georgia
| | - Mark Krasnow
- Stanford University School of Medicine, Stanford, California
| | - James Lee
- The Ohio State University, Columbus, Ohio
| | - Se-Hoon Lee
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | | | - Benjamin Lok
- Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | | | | | | | - John Minna
- UT Southwestern Medical Center, Dallas, Texas
| | - Matthew Oser
- Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Keunchil Park
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | | | - Yves Pommier
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | | | | | - Julien Sage
- Stanford University School of Medicine, Stanford, California
| | | | - Martin L Sos
- Department of Translational Genomics, Center of Integrated Oncology Cologne-Bonn, Medical Faculty, University of Cologne, Cologne Germany; Molecular Pathology, Institute of Pathology, University Hospital Cologne, Cologne, Germany; Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Kate D Sutherland
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | | | | | - Sarah J Wait
- Huntsman Cancer Institute and University of Utah, Salt Lake City, Utah
| | | | - Kwok Kin Wong
- Perlmutter Cancer Center, New York University Langone Health, New York, New York
| | - Hua Zhang
- Perlmutter Cancer Center, New York University Langone Health, New York, New York
| | - Jillian Daigneault
- International Association for the Study of Lung Cancer, Aurora, Colorado
| | - Jacinta Wiens
- International Association for the Study of Lung Cancer, Aurora, Colorado
| | | | - Trudy G Oliver
- Huntsman Cancer Institute and University of Utah, Salt Lake City, Utah.
| |
Collapse
|
46
|
New opportunities in a challenging disease: lurbinectedin for relapsed small-cell lung cancer. Lancet Oncol 2020; 21:605-607. [PMID: 32224305 DOI: 10.1016/s1470-2045(20)30097-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 02/10/2020] [Accepted: 02/11/2020] [Indexed: 10/24/2022]
|
47
|
Kauffmann-Guerrero D, Huber RM. Orphan Drugs in Development for the Treatment of Small-Cell Lung Cancer: Emerging Data on Lurbinectedin. LUNG CANCER-TARGETS AND THERAPY 2020; 11:27-31. [PMID: 32184690 PMCID: PMC7060031 DOI: 10.2147/lctt.s239223] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 02/07/2020] [Indexed: 01/05/2023]
Abstract
Lung cancer is the leading cause of death of all cancer entities and small-cell lung cancer (SCLC) is the most malignant subtype. Despite good initial response to chemotherapy, many patients relapse early and success of second line treatment remains poor. For years, no relevant improvement of second line treatment has been achieved in the field of SCLC. Lurbinectedin, a novel RNA-polymerase II inhibitor has shown promising results in pretreated SCLC patients as single agent and in combination with other chemotherapeutic drugs leading to an orphan drug designation from the FDA. This article reviews the current data on this emerging substance and its impact on the treatment of SCLC.
Collapse
Affiliation(s)
- Diego Kauffmann-Guerrero
- Division of Respiratory Medicine and Thoracic Oncology, Department of Internal Medicine V and Thoracic Oncology Centre Munich, University of Munich (LMU), Comprehensive Pneumology Center, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Rudolf Maria Huber
- Division of Respiratory Medicine and Thoracic Oncology, Department of Internal Medicine V and Thoracic Oncology Centre Munich, University of Munich (LMU), Comprehensive Pneumology Center, Member of the German Center for Lung Research (DZL), Munich, Germany
| |
Collapse
|
48
|
Guo H, Li L, Cui J. Advances and challenges in immunotherapy of small cell lung cancer. Chin J Cancer Res 2020; 32:115-128. [PMID: 32194311 PMCID: PMC7072020 DOI: 10.21147/j.issn.1000-9604.2020.01.13] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 12/31/2019] [Indexed: 12/19/2022] Open
Abstract
Small cell lung cancer (SCLC) is a highly lethal disease, characterized by early metastasis and rapid growth, and no effective treatment after relapse. Etoposide-platinum (EP) combination has been the backbone therapy of SCLC over the past 30 years. It is extremely urgent and important to seek new therapies for SCLC. In the past 5 years, immunotherapy, such as immune checkpoint inhibitors programmed cell death protein-1 (PD-1), cytotoxic T lymphocyte associatedprotein-4 (CTLA-4), has made remarkable achievements in the treatment of patients with SCLC, and it has become the first-line option for the treatment of some patients. Some traditional chemotherapeutic drugs or targeted drugs, such as alkylating agent temozolomide and transcription inhibitor lurbinectedin, have been found to have immunomodulatory effects and are expected to become new immunotherapeutic agents. In this study, we aimed to review the efficacy of new treatments for SCLC and discuss the current challenges and application prospect in the treatment of SCLC patients.
Collapse
Affiliation(s)
- Hanfei Guo
- Cancer Center, The First Hospital of Jilin University, Changchun 130021, China
| | - Lingyu Li
- Cancer Center, The First Hospital of Jilin University, Changchun 130021, China
| | - Jiuwei Cui
- Cancer Center, The First Hospital of Jilin University, Changchun 130021, China
| |
Collapse
|
49
|
Jimenez PC, Wilke DV, Branco PC, Bauermeister A, Rezende‐Teixeira P, Gaudêncio SP, Costa‐Lotufo LV. Enriching cancer pharmacology with drugs of marine origin. Br J Pharmacol 2020; 177:3-27. [PMID: 31621891 PMCID: PMC6976878 DOI: 10.1111/bph.14876] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 08/13/2019] [Accepted: 09/05/2019] [Indexed: 12/29/2022] Open
Abstract
Marine natural products have proven, over the last half-century, to be effective biological modulators. These molecules have revealed new targets for cancer therapy as well as dissimilar modes of action within typical classes of drugs. In this scenario, innovation from marine-based pharmaceuticals has helped advance cancer chemotherapy in many aspects, as most of these are designated as first-in-class drugs. Here, by examining the path from discovery to development of clinically approved drugs of marine origin for cancer treatment-cytarabine (Cytosar-U®), trabectedin (Yondelis®), eribulin (Halaven®), brentuximab vedotin (Adcetris®), and plitidepsin (Aplidin®)- together with those in late clinical trial phases-lurbinectedin, plinabulin, marizomib, and plocabulin-the present review offers a critical analysis of the contributions given by these new compounds to cancer pharmacotherapy.
Collapse
Affiliation(s)
- Paula C. Jimenez
- Departamento de Ciências do MarUniversidade Federal de São PauloSantosSPBrasil
| | - Diego V. Wilke
- Núcleo de Pesquisa e Desenvolvimento de Medicamentos (NPDM), Departamento de Fisiologia e Farmacologia, Faculdade de MedicinaUniversidade Federal do CearáFortalezaCEBrasil
| | - Paola C. Branco
- Departamento de Farmacologia, Instituto de Ciências BiomédicasUniversidade de São PauloSão PauloSPBrasil
| | - Anelize Bauermeister
- Departamento de Farmacologia, Instituto de Ciências BiomédicasUniversidade de São PauloSão PauloSPBrasil
| | - Paula Rezende‐Teixeira
- Departamento de Farmacologia, Instituto de Ciências BiomédicasUniversidade de São PauloSão PauloSPBrasil
| | - Susana P. Gaudêncio
- UCIBIO, Department of Chemistry, Blue Biotechnology and Biomedicine Lab, Faculty of Science and TechnologyNOVA University of LisbonCaparicaPortugal
| | - Leticia V. Costa‐Lotufo
- Departamento de Farmacologia, Instituto de Ciências BiomédicasUniversidade de São PauloSão PauloSPBrasil
| |
Collapse
|
50
|
Cote GM, Choy E, Chen T, Marino-Enriquez A, Morgan J, Merriam P, Thornton K, Wagner AJ, Nathenson MJ, Demetri G, George S. A phase II multi-strata study of lurbinectedin as a single agent or in combination with conventional chemotherapy in metastatic and/or unresectable sarcomas. Eur J Cancer 2019; 126:21-32. [PMID: 31896519 DOI: 10.1016/j.ejca.2019.10.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 10/18/2019] [Accepted: 10/21/2019] [Indexed: 01/22/2023]
Abstract
Chemotherapy objective response rates (ORRs) in metastatic soft tissue sarcoma (STS) are typically 20-40% with median progression-free survival (PFS) less than 6 months. Lurbinectedin is a new anticancer agent under investigation. The primary objective of this three-arm, phase II study was to determine the disease control rate (DCR = ORR + stable disease [SD]) at 24 weeks of lurbinectedin alone or with chemotherapy in STS. Eligible patients included adults with ≤2 prior cytotoxic therapies. Study cohorts were: stratum A (StrA; anthracycline-naive), lurbinectedin/doxorubicin; stratum B (StrB; prior anthracycline), lurbinectedin/gemcitabine; stratum C (StrC; prior anthracycline/gemcitabine) lurbinectedin monotherapy. Each stratum was analysed separately by Simon two-stage design. Forty-two patients were accrued (StrA = 20, StrB = 10, StrC = 12) including leiomyosarcoma [LMS] (n = 20), synovial sarcoma [SS](n = 4), malignant peripheral nerve sheath tumour (n = 3) and other STS histologies (n = 15). For StrA there were seven partial responses (PR) plus one stable disease (SD) at 24 weeks. For StrB, two patients met the 24-week DCR including one PR (leiomyosarcoma) and one SD (desmoplastic small round cell tumour [DSRCT]). StrB did not continue to the second stage. In StrC, no patients met the primary end-point. Median progression-free survival (PFS) was: StrA = 4.2 months (90% CI 1.4-7.8), StrB = 1.7 months (90% confidence interval (CI) 1.0-7.4), and StrC = 1.3 months (90% CI 1.1-3.0). Lurbinectedin as a single agent or with chemotherapy was well tolerated with haematologic adverse events (AE's) as the most common toxicity. There were no treatment-related deaths. The combination of lurbinectedin/doxorubicin reached the DCR end-point with seven PR and one patient with SD (ORR 35.0%, 24-week DCR 40.0%). Evidence of drug benefit was seen in leiomyosarcoma, dedifferentiated liposarcoma (DDLS), myxoid liposarcoma (MLS), synovial sarcoma (SS), and desmoplastic small round cell tumour (DSRCT). TRIAL REGISTRATION: clinicaltrials.gov; NCT02448537.
Collapse
Affiliation(s)
- Gregory M Cote
- Division of Hematology and Oncology, Department of Medicine, Boston, MA, USA.
| | - Edwin Choy
- Division of Hematology and Oncology, Department of Medicine, Boston, MA, USA
| | - Tianqi Chen
- Department of Data Sciences, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Adrian Marino-Enriquez
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Jeffrey Morgan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Priscilla Merriam
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Katherine Thornton
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Andrew J Wagner
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Michael J Nathenson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - George Demetri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Suzanne George
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|