1
|
Kim SY, Park HS, Chiang AC. Small Cell Lung Cancer: A Review. JAMA 2025:2832148. [PMID: 40163214 DOI: 10.1001/jama.2025.0560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Importance Small cell lung cancer (SCLC) is a high-grade neuroendocrine carcinoma with an incidence of 4.7 cases per 100 000 individuals in 2021 in the US and a 5-year overall survival of 12% to 30%. Observations Cigarette smoking is the primary risk factor for development of SCLC, as 95% of patients diagnosed with SCLC have a history of tobacco use. Patients with SCLC may present with respiratory symptoms such as cough (40%), shortness of breath (34%), hemoptysis (10%), or metastases with corresponding local symptoms (30%) such as pleuritis or bone pain; approximately 60% of patients with SCLC may be asymptomatic at diagnosis. Chest imaging may demonstrate central hilar (85%) or mediastinal lymphadenopathy (75%). At diagnosis, approximately 15% of patients have brain metastases, which may present as headache or focal weakness. Diagnosis is confirmed by biopsy of a primary lung mass, thoracic lymph node, or metastatic lesion. Small cell lung cancer is classified into limited stage (LS-SCLC; 30%) vs extensive stage (ES-SCLC; 70%) based on whether the disease can be treated within a radiation field that is typically confined to 1 hemithorax but may include contralateral mediastinal and supraclavicular nodes. For patients with LS-SCLC, surgery or concurrent chemotherapy with platinum-etoposide and radiotherapy is potentially curative in 30% of patients. More recently, median survival for LS-SCLC has reached up to 55.9 months with the addition of durvalumab, an immunotherapy. First-line treatment for ES-SCLC is combined treatment with platinum-etoposide chemotherapy and immunotherapy with the programmed cell death 1 ligand 1 (PD-L1) inhibitors durvalumab or atezolizumab followed by maintenance immunotherapy until disease progression or toxicity. Although initial rates of tumor shrinkage are 60% to 70% with platinum-etoposide and immunotherapy treatment, the median overall survival of patients treated for ES-SCLC is approximately 12 to 13 months, with 60% of patients relapsing within 3 months. Second-line therapy for patients with ES-SCLC includes the DNA-alkylating agent lurbinectedin (35% overall response rate; median progression-free survival, 3.7 months) and a bispecific T-cell engager against delta-like ligand 3, tarlatamab (40% overall response rate; median progression-free survival, 4.9 months). Conclusions and Relevance Small cell lung cancer is a smoking-related malignancy that presents at an advanced stage in 70% of patients. Three-year overall survival is approximately 56.5% for LS-SCLC and 17.6% for ES-SCLC. First-line treatment for LS-SCLC is radiation targeting the tumor given concurrently with chemotherapy and followed by consolidation immunotherapy. For ES-SCLC, first-line treatment is chemotherapy and immunotherapy followed by maintenance immunotherapy.
Collapse
Affiliation(s)
- So Yeon Kim
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Henry S Park
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, Connecticut
| | - Anne C Chiang
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
2
|
Li S, Lv J, Li Z, Zhang Q, Lu J, Huo X, Guo M, Liu X, Li C, Wang J, Shi H, Deng L, Chen Z, Du X. Overcoming multi-drug resistance in SCLC: a synergistic approach with venetoclax and hydroxychloroquine targeting the lncRNA LYPLAL1-DT/BCL2/BECN1 pathway. Mol Cancer 2024; 23:243. [PMID: 39478582 PMCID: PMC11526623 DOI: 10.1186/s12943-024-02145-1] [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: 04/26/2024] [Accepted: 10/04/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND Small cell lung cancer (SCLC) stands as one of the most lethal malignancies, characterized by a grim diagnosis and prognosis. The emergence of multi-drug resistance poses a significant hurdle to effective therapy. Although previous studies have implicated the long noncoding RNA LYPLAL1-DT in the tumorigenesis of SCLC, the precise role of the highly expressed LYPLAL1-DT in SCLC chemoresistance and the underlying mechanism remain inadequately understood. METHODS cDDP-, VP-16- and PTX-resistant SCLC cells lines were established. The viabilities of SCLC cells were assessed by CCK-8 assay in vitro and xenograft tumor formation assay in vivo. Apoptosis was evaluated by FACS, Western blot and JC-1 fluorescence staining, while autophagy was explored via autophagic flux detection under confocal microscopy and autophagic vacuole investigation under transmission electron microscopy (TEM). The functional role and mechanism of LYPLAL1-DT were further investigated by gain- and loss-of-function assays in vitro. Furthermore, the therapeutic efficacy of the combination of venetoclax and HCQ with cDDP, VP-16 or PTX was evaluated by cell line, cell-derived xenograft (CDX) and patient-derived xenograft (PDX) mice model. RESULTS Our findings revealed that LYPLAL1-DT is upregulated in chemoresistant SCLC cell lines. Gain- and loss-of-function assays demonstrated that LYPLAL1-DT impairs sensitivity to cDDP, VP-16, or PTX both in vitro and in vivo. Overexpression of LYPLAL1-DT significantly enhanced autophagy and inhibited apoptosis in SCLC cells. Further analyses, including RIP and RNA pull-down assays, revealed that LYPLAL1-DT promotes the expression of BCL2 by sponging miR-204-5p and is implicated in the assembly of the autophagy-specific complex (BECN1/PtdIns3K complex). Combining venetoclax and HCQ with cDDP, VP-16, or PTX effectively mitigated chemoresistance in SCLC cells and suppressed tumor growth in CDX and PDX models without inducing obvious toxic effects. CONCLUSIONS Our findings demonstrate that upregulation of LYPLAL1-DT sequesters apoptosis through the LYPLAL1-DT/miR-204-5p/BCL2 axis and promotes autophagy by facilitating the assembly of the BECN1/PtdIns3K complex, thereby mediating multi-drug resistance of SCLC. The triple combination of venetoclax, HCQ, in conjunction with cDDP, VP-16 or PTX overcomes refractory SCLC, shedding light on a potential therapeutic target for combating SCLC chemoresistance.
Collapse
Affiliation(s)
- Shuxin Li
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, 100069, China
| | - Jianyi Lv
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, 100069, China
| | - Zhihui Li
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, 100069, China
| | - Qiuyu Zhang
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, 100069, China
| | - Jing Lu
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, 100069, China
| | - Xueyun Huo
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, 100069, China
| | - Meng Guo
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, 100069, China
| | - Xin Liu
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, 100069, China
| | - Changlong Li
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, 100069, China
| | - Jinghui Wang
- Department of Medical Oncology, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, Beijing, 101149, China
| | - Hanping Shi
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100069, China
- Department of Gastrointestinal Surgery/Clinical Nutrition, Capital Medical University Affiliated Beijing Shijitan Hospital, Beijing, 100038, China
| | - Li Deng
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100069, China
- Department of Gastrointestinal Surgery/Clinical Nutrition, Capital Medical University Affiliated Beijing Shijitan Hospital, Beijing, 100038, China
| | - Zhenwen Chen
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, 100069, China
| | - Xiaoyan Du
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China.
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100069, China.
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, 100069, China.
| |
Collapse
|
3
|
Nie Q, Chen W, Zhang T, Ye S, Ren Z, Zhang P, Wen J. Iron oxide nanoparticles induce ferroptosis via the autophagic pathway by synergistic bundling with paclitaxel. Mol Med Rep 2023; 28:198. [PMID: 37681444 PMCID: PMC10510030 DOI: 10.3892/mmr.2023.13085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 08/08/2023] [Indexed: 09/09/2023] Open
Abstract
In recent years, inhibiting tumor cell activity by triggering cell ferroptosis has become a research hotspot. The development of generic targeted nanotherapeutics might bring new ideas for non‑invasive applications. Currently, the potential mechanism underlying the universal application of paclitaxel (PTX)‑loaded iron oxide nanoparticles (IONP@PTX) to different types of tumors is unclear. The present study aimed to prepare IONP@PTX for targeted cancer therapy and further explore the potential mechanisms underlying the inhibitory effects of this material on the NCI‑H446 human small cell lung cancer and brain M059K malignant glioblastoma cell lines. First, a CCK‑8 assay was performed to determine cell viability, and then the combination index for evaluating drug combination interaction effect was evaluated. Intracellular reactive oxygen species (ROS) and lipid peroxidation levels were monitored using a DCFH‑DA fluorescent probe and a C11‑BODIPY™ fluorescent probe, respectively. Furthermore, western blotting assay was performed to determine the expression of autophagy‑ and iron death‑related proteins. The experimental results showed that, compared with either IONP monotherapy, PTX monotherapy, or IONP + PTX, IONP@PTX exerted a synergistic effect on the viability of both cell types, with significantly increased total iron ion concentration, ROS levels and lipid peroxidation levels. IONP@PTX significantly increased the expression of autophagy‑related proteins Beclin 1 and histone deacetylase 6 (HDAC6) in both cell lines (P<0.05), increased the expression of light chain 3 (LC3)‑II/I in NCI‑H446 cells (P<0.05) and decreased that of sequestosome1 (p62) in M059K cells (P<0.05). Moreover, the addition of rapamycin enhanced the IONP@PTX‑induced the upregulation of Beclin 1, LC3‑II/I and HDAC6 and the downregulation of mTORC1 protein in both cell lines (P<0.05). Moreover, rapamycin enhanced the IONP@PTX‑induced downregulation of p62 protein in NCI‑H446 cells (P<0.05), suggesting that IONP@PTX induces ferroptosis, most likely through autophagy. Collectively, the present findings show that IONP works synergistically with PTX to induce ferroptosis via the autophagic pathway.
Collapse
Affiliation(s)
- Qi Nie
- Guangxi Clinical Medical Research Center for Neurological Diseases, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, P.R. China
- College of Pharmacy, Guilin Medical University, Guilin, Guangxi 541104, P.R. China
| | - Wenqing Chen
- Guangxi Clinical Medical Research Center for Neurological Diseases, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, P.R. China
- College of Pharmacy, Guilin Medical University, Guilin, Guangxi 541104, P.R. China
| | - Tianmei Zhang
- Guangxi Clinical Medical Research Center for Neurological Diseases, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, P.R. China
| | - Shangrong Ye
- Guangxi Clinical Medical Research Center for Neurological Diseases, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, P.R. China
| | - Zhongyu Ren
- Guangxi Clinical Medical Research Center for Neurological Diseases, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, P.R. China
| | - Peng Zhang
- Guangxi Clinical Medical Research Center for Neurological Diseases, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, P.R. China
| | - Jian Wen
- Guangxi Clinical Medical Research Center for Neurological Diseases, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, P.R. China
- College of Pharmacy, Guilin Medical University, Guilin, Guangxi 541104, P.R. China
| |
Collapse
|
4
|
Mu F, Fan B, Li H, Qin W, Wang C, Zou B, Wang L. Impact of nab-paclitaxel plus PD-1/PD-L1 inhibitor on chemorefractory relapsed small-cell lung cancer. Future Oncol 2023. [PMID: 37114967 DOI: 10.2217/fon-2023-0076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2023] Open
Abstract
Background: The present study evaluated the efficacy and safety of nab-paclitaxel (nab-PTX) with a concurrent PD-1/PD-L1 inhibitor in patients with refractory relapsed small-cell lung cancer (SCLC). Patients & methods: We retrospectively analyzed 240 patients with refractory relapsed SCLC: 40 patients were treated with nab-PTX plus PD-1/PD-L1 inhibitor, and 200 received traditional chemotherapy. Results: Median progression-free survival in the nab-PTX plus PD-1/PD-L1 inhibitor and traditional chemotherapy groups was 3.6 and 2.5 months (p = 0.0021), respectively. The median overall survival was 8.0 and 5.2 months (p = 0.0002), respectively. No new safety issues were identified. Conclusion: Nab-PTX plus PD-1/PD-L1 inhibitor significantly improved survival in patients with refractory relapsed SCLC compared with traditional chemotherapy.
Collapse
Affiliation(s)
- Fengchun Mu
- Department of Radiation Oncology, Shandong Cancer Hospital & Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
| | - Bingjie Fan
- Department of Radiation Oncology, Shandong Cancer Hospital & Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
| | - Haoqian Li
- Department of Radiation Oncology, Shandong Cancer Hospital & Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
| | - Wenru Qin
- Department of Radiation Oncology, Shandong Cancer Hospital & Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
| | - Chunni Wang
- Department of Radiation Oncology, Shandong Cancer Hospital & Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
| | - Bing Zou
- Department of Radiation Oncology, Shandong Cancer Hospital & Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
| | - Linlin Wang
- Department of Radiation Oncology, Shandong Cancer Hospital & Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
| |
Collapse
|
5
|
Gong C, Xie Y, Zhao Y, Li Y, Zhang J, Wang L, Cao J, Tao Z, Hu X, Wang B. Comparison of two regimens of weekly paclitaxel plus gemcitabine in patients with metastatic breast cancer: propensity score-matched analysis of real-world data. Ther Adv Drug Saf 2022; 13:20420986221146411. [PMID: 36582188 PMCID: PMC9793024 DOI: 10.1177/20420986221146411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 12/01/2022] [Indexed: 12/25/2022] Open
Abstract
Purpose Weekly gemcitabine + paclitaxel (wGT) administration is widely applied in real-world clinical practice. The 28-day and 21-day regimens of wGT are the most widely accepted regimens. We evaluated the efficacy and safety of wGT administration in patients with metastatic breast cancer (MBC) and compared the two regimens. Methods Patients with human epidermal growth factor receptor 2 (HER-2)-negative MBC who received wGT between October 2013 and October 2016 were identified using an electronic database. The outcome variables included progression-free survival (PFS), overall survival (OS), objective response rate (ORR), and safety profile. Propensity score matching was performed to minimize potential confounders. Results A total of 140 patients were included. The median PFS and OS was 7.8 [95% confidence interval (CI) = 7.0-8.7] months and 22.5 (95% CI = 18.8-26.1) months, respectively. The toxicity of wGT was manageable. Among the patients, 90 (64.3%) received the 21-day regimen and 50 (35.7%) received the 28-day regimen. A higher number of younger patients and patients receiving later-line therapy received the 28-day regimen. There was no significant difference between the two groups in PFS after propensity score matching, though subgroup analysis showed that patients with early relapse benefited more from the 28-day regimen. The ORR was numerically higher in 28-day regimen (37.8% versus 28.0%, p = 0.310). However, the 21-day regimen was better tolerated than the 28-day regimen. Conclusion wGT administration showed efficacy and safety in patients with MBC. The efficacy was comparable between the two regimens after adjustment for confounding factors while the 21-day regimen was better tolerated. Plain Language Summary 21-day regimen of wGT was well tolerated in patients with metastatic breast cancer Weekly gemcitabine + paclitaxel (wGT) administration is widely applied in real-world clinical practice. The 28-day and 21-day regimens of wGT are the most widely accepted regimens. We evaluated the efficacy and safety of wGT administration in patients with metastatic breast cancer (MBC) and compared the two regimens. Patients with human epidermal growth factor receptor 2 (HER-2)-negative MBC who received wGT between October 2013 and October 2016 were identified using an electronic database. The outcome variables included progression-free survival (PFS), overall survival (OS), objective response rate (ORR), and safety profile. Propensity score matching was performed to minimize potential confounders. We found that the efficacy was comparable between the two regimens after adjustment for confounding factors while the 21-day regimen was better tolerated.
Collapse
Affiliation(s)
| | | | - Yannan Zhao
- Department of Breast and Urological Medical
Oncology, Fudan University Shanghai Cancer Center, Shanghai, China,Department of Oncology, Shanghai Medical
College, Fudan University, Shanghai, China
| | - Yi Li
- Department of Breast and Urological Medical
Oncology, Fudan University Shanghai Cancer Center, Shanghai, China,Department of Oncology, Shanghai Medical
College, Fudan University, Shanghai, China
| | - Jian Zhang
- Department of Breast and Urological Medical
Oncology, Fudan University Shanghai Cancer Center, Shanghai, China,Department of Oncology, Shanghai Medical
College, Fudan University, Shanghai, China
| | - Leiping Wang
- Department of Breast and Urological Medical
Oncology, Fudan University Shanghai Cancer Center, Shanghai, China,Department of Oncology, Shanghai Medical
College, Fudan University, Shanghai, China
| | - Jun Cao
- Department of Breast and Urological Medical
Oncology, Fudan University Shanghai Cancer Center, Shanghai, China,Department of Oncology, Shanghai Medical
College, Fudan University, Shanghai, China
| | - Zhonghua Tao
- Department of Breast and Urological Medical
Oncology, Fudan University Shanghai Cancer Center, Shanghai, China,Department of Oncology, Shanghai Medical
College, Fudan University, Shanghai, China
| | - Xichun Hu
- Department of Breast and Urological Medical
Oncology, Fudan University Shanghai Cancer Center, Shanghai, China,Department of Oncology, Shanghai Medical
College, Fudan University, Shanghai 200032, China
| | | |
Collapse
|
6
|
Hameedat F, Pizarroso NA, Teixeira N, Pinto S, Sarmento B. Functionalized FcRn-targeted nanosystems for oral drug delivery: A new approach to colorectal cancer treatment. Eur J Pharm Sci 2022; 176:106259. [PMID: 35842140 DOI: 10.1016/j.ejps.2022.106259] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 07/01/2022] [Accepted: 07/11/2022] [Indexed: 01/17/2023]
Abstract
Colorectal cancer (CRC) is the second type of cancer with the highest lethality rate. The current chemotherapy to treat CRC causes systemic toxicity, unsatisfying response rate, and low tumor-specific selectivity, which is mainly administered by invasive routes. The chronic and aggressive nature of cancers may require long-term regimens. Thus, the oral route is preferred. However, the orally administered drugs still need to surpass the harsh environment of the gastrointestinal tract and the biological barriers. Nanotechnology is a promising strategy to overcome the oral route limitations. Targeted nanoparticle systems decorated with functional groups can enhance the delivery of anticancer agents to tumor sites. It is described in the literature that the neonatal Fc receptor (FcRn) is expressed in cancer tissue and overexpressed in CRC epithelial cells. However, the impact of FcRn-targeted nanosystems in the treatment of CRC has been poorly investigated. This review article discusses the current knowledge on the involvement of the FcRn in CRC, as well as to critically assess its relevance as a target for further localization of oral nanocarriers in CRC tumor cells. Finally, a brief overview of cancer therapeutics, strategies to design the nanoparticles of anticancer drugs and a review of decorated nanoparticles with FcRn moieties are explored.
Collapse
Affiliation(s)
- Fatima Hameedat
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto 4200-393, Portugal; NANOMED EMJMD, Pharmacy School, Faculty of Health, University of Angers, France; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, Porto 4200-393, Portugal
| | - Nuria A Pizarroso
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto 4200-393, Portugal
| | - Natália Teixeira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto 4200-393, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, Porto 4200-393, Portugal; Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Rua do Campo Alegre, Porto 4169-007, Portugal
| | - Soraia Pinto
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto 4200-393, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, Porto 4200-393, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Rua Jorge Viterbo Ferreira, 228, Porto 4150-180, Portugal
| | - Bruno Sarmento
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto 4200-393, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, Porto 4200-393, Portugal; CESPU - IUCS, Rua Central de Gandra 1317, Gandra 4585-116, Portugal.
| |
Collapse
|
7
|
Stratmann JA, Timalsina R, Atmaca A, Rosery V, Frost N, Alt J, Waller CF, Reinmuth N, Rohde G, Saalfeld FC, von Rose AB, Acker F, Aspacher L, Möller M, Sebastian M. Clinical predictors of survival in patients with relapsed/refractory small-cell lung cancer treated with checkpoint inhibitors: a German multicentric real-world analysis. Ther Adv Med Oncol 2022; 14:17588359221097191. [PMID: 35677321 PMCID: PMC9168937 DOI: 10.1177/17588359221097191] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 04/11/2022] [Indexed: 01/22/2023] Open
Abstract
Objectives: Small-cell lung cancer (SCLC) is a lung malignancy with high relapse rates
and poor survival outcomes. Treatment-resistant disease relapse occurs
frequently and effective salvage therapies are urgently needed. Materials and Methods: We aimed to define efficacy and safety of checkpoint inhibitors (CPIs) in a
heterogeneous population of relapsed and refractory SCLC patients in a large
retrospective multicentric real-world cohort across German tertiary care
centers. Results: A total of 111 patients from 11 treatment centers were included. Median age
of all patients was 64 years, and 63% were male. Approximately one-third of
all patients had poor performance status [Eastern Cooperative Oncology Group
(ECOG) ⩾ 2], and 37% had known brain metastases. Patients were heavily
pretreated with a median number of prior therapy lines of 2 (range, 1–8).
Median follow-up of the entire cohort was 21.7 months. Nivolumab and
Nivolumab/Ipilimumab were the most common regimens. Overall disease control
rate was 27.2% in all patients and was numerically higher in CPI combination
regimens compared with single-agent CPI (31.8% versus
23.8%; p = 0.16). Median overall survival (OS) was
5.8 months [95% confidence interval (CI), 1.7–9.9 months]. The 12- and
24-month survival rates were 31.8% and 12.7%, respectively. The 12-week
death rate was 27.9%. Disease control and response rate were significantly
lower in patients with liver metastases. Platinum sensitivity (to first-line
treatment), metastatic burden, and lactate dehydrogenase (LDH) showed
prognostic impact on survival in univariate analysis.
Neutrophil-to-lymphocyte ratio (NLR) was a significant and independent
predictor of survival in univariate (p = 0.01) and
multivariate analyses [hazard ratio (HR), 2.1; 95% CI = 1.1–4.1;
p = 0.03]. Conclusion: CPI in patients with relapsed or refractory (R/R) SCLC is of limited value in
an overall patient cohort; however, long-term survival, in particular with
CPI combination strategies, is possible. Clinical characteristics allow a
more differentiated subgroup selection, in particular patients with low NLR
showed less benefit from CPI in R/R SCLC.
Collapse
Affiliation(s)
- Jan A. Stratmann
- Department of Internal Medicine, Hematology/Oncology, Goethe University, Theodor Stern Kai 7, 60596 Frankfurt am Main, Germany
| | - Radha Timalsina
- Department of Internal Medicine, Hematology/Oncology, Goethe University, Frankfurt, Germany
| | - Akin Atmaca
- Department of Oncology and Hematology, Krankenhaus Nordwest, UCT-University Cancer Center, Frankfurt, Germany
| | - Vivian Rosery
- Department of Medical Oncology, West German Cancer Center, University Medicine Essen, Essen, Germany
| | - Nikolaj Frost
- Charité – Universitätsmedizin Berlin, Berlin, GermanyHumboldt-Universität zu Berlin, Berlin, Germany
- Department of Infectious Diseases and Pulmonary Medicine, Berlin Institute of Health, Berlin, Germany
| | - Jürgen Alt
- Department of Internal Medicine III (Hematology, Oncology, Pneumology), University Medical Center Mainz, Mainz, Germany
| | - Cornelius F. Waller
- Internal Medicine I, Haematology, Oncology and Stem Cell Transplantation, Faculty of Medicine, Freiburg University Medical Center, Freiburg, Germany
| | - Niels Reinmuth
- Department of Oncology, Asklepios Clinic München-Gauting, Gauting, Germany
| | - Gernot Rohde
- Department of Respiratory Medicine, Medical Clinic 1, University Hospital, Frankfurt, Germany
| | - Felix C. Saalfeld
- Department for Internal Medicine I, University Hospital Carl Gustav Carus Dresden, TU Dresden, Dresden, Germany
| | - Aaron Becker von Rose
- Department of Internal Medicine III, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Fabian Acker
- Department of Internal Medicine, Hematology/Oncology, Goethe University, Frankfurt, Germany
| | - Lukas Aspacher
- Department of Internal Medicine, Hematology/Oncology, Goethe University, Frankfurt, Germany
| | - Miriam Möller
- Department of Oncology, Martha–Maria Hospital Halle, Halle, Germany
| | - Martin Sebastian
- Department of Internal Medicine, Hematology/Oncology, Goethe University, Frankfurt, Germany
| |
Collapse
|
8
|
Hao Z, Sekkath Veedu J. Current Strategies for Extensive Stage Small Cell Lung Cancer Beyond First-line Therapy. Clin Lung Cancer 2021; 23:14-20. [PMID: 34656433 DOI: 10.1016/j.cllc.2021.09.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 08/22/2021] [Accepted: 09/09/2021] [Indexed: 02/07/2023]
Abstract
Extensive stage small cell lung cancer carries extremely poor prognosis and adding immune checkpoint inhibitor to platinum etoposide combination in first line only improved outcomes modestly. Once disease recurs, treatment response is only transient in nature. Various strategies that are being explored include dual checkpoint blockade, BiTE and CAR-T cell approaches. Immune checkpoint inhibitors are being combined with PARP inhibitors. Other approaches currently being investigated include liposomal irinotecan and combining known active agents for SCLC in relapsed setting such as newly approved lurbinectedin with doxorubicin, paclitaxel, irinotecan or topotecan with ATR inhibitor (Berzosertib). Temozolomide has also been tested in combination with a Parp inhibitor. New antibody or small molecule drug conjugates are being actively investigated, so is a biomarker based approach. Better understanding of small cell lung cancer disease biology via high through-put genomic, proteomic and methylation profiling offer glimpse of hope in our efforts to contain this deadly disease. A table of representative molecular targets under investigation is provided in the end.
Collapse
Affiliation(s)
- Zhonglin Hao
- Division of Medical Oncology, Department of Medicine, Markey Cancer Center, College of Medicine, University of Kentucky, Lexington KY.
| | - Janeesh Sekkath Veedu
- Division of Medical Oncology, Department of Medicine, Markey Cancer Center, College of Medicine, University of Kentucky, Lexington KY
| |
Collapse
|
9
|
Rudnik-Jansen I, Howard KA. FcRn expression in cancer: Mechanistic basis and therapeutic opportunities. J Control Release 2021; 337:248-257. [PMID: 34245786 DOI: 10.1016/j.jconrel.2021.07.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 07/04/2021] [Accepted: 07/05/2021] [Indexed: 01/30/2023]
Abstract
There is an urgent need to identify new cellular targets to expand the repertoire, potency and safety of cancer therapeutics. Neonatal Fc Receptor (FcRn)-driven cellular recycling plays a predominant role in the prolonged serum half-life of human serum albumin (HSA) and immunoglobulin G (IgG) exploited in long-acting cancer drug designs. FcRn-mediated HSA and IgG uptake in epithelial cells and dendritic cell antigen presentation offers new therapeutic opportunities beyond half-life extension. Altered FcRn expression in solid tumours accounting for HSA catabolism or recycling supports a role for FcRn in tumour metabolism and growth. This review addresses the mechanistic basis for different FcRn expression profiles observed in cancer and exploitation for targeted drug delivery. Furthermore, the review highlights FcRn-mediated immunosurveillance and immune therapy. FcRn offers a potential attractive cancer target but in-depth understanding of role and expression profiles during cancer pathogenesis is required for tailoring targeted drug designs.
Collapse
Affiliation(s)
- Imke Rudnik-Jansen
- Interdisciplinary Nanoscience Center (iNANO), Department of Molecular Biology and Genetics, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Kenneth A Howard
- Interdisciplinary Nanoscience Center (iNANO), Department of Molecular Biology and Genetics, Aarhus University, DK-8000 Aarhus C, Denmark.
| |
Collapse
|
10
|
Wang Y, Li L, Xu C. Nanoparticle Albumin Bound Paclitaxel in the Third-Line Treatment of Recurrent Small Cell Lung Cancer in Real-World Practice: A Single Center Experience. Technol Cancer Res Treat 2021; 20:15330338211050775. [PMID: 34657492 PMCID: PMC8642115 DOI: 10.1177/15330338211050775] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 08/25/2021] [Accepted: 09/16/2021] [Indexed: 11/19/2022] Open
Abstract
Background: Small cell lung cancer (SCLC) is a type of highly malignant tumor. It is easy to relapse and high resistant to second-line chemotherapy. There is not a standard plan for third-line and subsequent-line treatment, hence the current study aimed to explore the performance of nanoparticle albumin bound paclitaxel (Nab-P) in the third-line treatment of recurrent SCLC. Methods: A retrospective analysis of pathologically confirmed third-line SCLC patients was conducted to observe the efficacy and adverse reactions of Nab-P treatment. Results: Among the 37 SCLC patients included in the study, objective response rate was 24.32%, and disease control rate was 81.08%. The main adverse reactions were gastrointestinal reactions, bone marrow suppression, muscle and joint aches and fatigue. Adverse reactions ≥ level 3 included leukopenia & neutropenia (24.32%), thrombocytopenia (5.41%) and anemia (2.70%). Conclusions: As a single-agent third-line treatment for patients with relapsed SCLC, Nab-P was effective and safe.
Collapse
Affiliation(s)
- Yuchao Wang
- Nanjing Chest Hospital, Nanjing, China
- Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| | - Li Li
- Nanjing Chest Hospital, Nanjing, China
- Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| | - Chunhua Xu
- Nanjing Chest Hospital, Nanjing, China
- Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
11
|
Combination therapy with carboplatin and paclitaxel for small cell lung cancer. Respir Investig 2018; 57:34-39. [PMID: 30528688 DOI: 10.1016/j.resinv.2018.09.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 08/23/2018] [Accepted: 09/05/2018] [Indexed: 11/22/2022]
Abstract
BACKGROUND Although small cell lung cancer (SCLC) is an aggressive cancer, few useful treatment options exist after relapse. Information concerning the efficacy and safety of carboplatin plus paclitaxel in patients with SCLC is limited. METHODS From April 2007 to October 2016, 318 patients with SCLC received chemotherapy at our institution. The medical records of patients treated with carboplatin and paclitaxel after first-line chemotherapy with platinum plus etoposide or irinotecan were retrospectively analyzed. The objectives were to investigate the frequency at which a carboplatin and paclitaxel regimen was administered to patients with SCLC in clinical practice, and to determine the response rate, progression-free survival (PFS), and tolerability of such agents. RESULTS A total of 24 (7.5%) patients (male, n = 21; female, n = 3; median age, 67 years; performance status, 0-1/≥2, 15/8 patients; limited/extensive disease, 6/15 patients; sensitive/refractory relapse, 3/21 patients) were treated with carboplatin plus paclitaxel. This regimen was chosen due to interstitial lung disease (ILD) (n = 17), radiation pneumonitis (n = 3), combination with palliative radiation therapy (n = 2), and the presence of other cancers (n = 2). The response rate was 33.3%, and the disease control rate was 62.5%. The median PFS and overall survival were 4.1 and 8.7 months, respectively. Grade 3/4 hematologic toxicities observed included neutropenia (54.2%), anemia (4.2%), and thrombocytopenia (8.3%). With the exception of grade 3 neuropathies (n = 2), non-hematologic toxicities were mild. No patients experienced an acute exacerbation of ILD. CONCLUSION A combination of carboplatin plus paclitaxel as second-line chemotherapy is effective and feasible in patients with SCLC, especially in those with ILD.
Collapse
|
12
|
Baize N, Monnet I, Greillier L, Quere G, Kerjouan M, Janicot H, Vergnenegre A, Auliac JB, Chouaid C. Second-line treatments of small-cell lung cancers. Expert Rev Anticancer Ther 2017; 17:1033-1043. [DOI: 10.1080/14737140.2017.1372198] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Nathalie Baize
- UTTIOM (Unité Transversale de Thérapeutiques Innovantes en Oncologie Médicale), CHU Angers, France
| | - Isabelle Monnet
- Department of Pulmonology, Centre Hospitalier Intercommunal de Créteil, Créteil, France
| | - Laurent Greillier
- Service d’Oncologie Multidisciplinaire et Innovations Thérapeutiques, AP-HM, Aix-Marseille Université, Marseille, France
| | - Gilles Quere
- Respiratory Disease Department, Brest University Brest, Brest, France
| | - Mallorie Kerjouan
- Respiratory Disease Department, Pontchaillou University Hospital, Rennes, France
| | - Henri Janicot
- Service de pneumologie, CHU Clermont-Ferrand, Clermont Ferrand, France
| | - Alain Vergnenegre
- UOTC (Unité d’Oncologie Thoracique et Cutanée), CHU Limoges, Limoges, France
| | | | - Christos Chouaid
- Department of Pulmonology, Centre Hospitalier Intercommunal de Créteil, Créteil, France
| |
Collapse
|
13
|
Takei Y, Takahashi Y, Machida S, Taneichi A, Takahashi S, Nagashima T, Morisawa H, Saga Y, Matsubara S, Fujiwara H. Response to and toxicity of gemcitabine for recurrent ovarian cancer according to number of previous chemotherapy regimens. J Obstet Gynaecol Res 2016; 43:358-364. [PMID: 27935223 DOI: 10.1111/jog.13203] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 09/04/2016] [Accepted: 09/11/2016] [Indexed: 11/30/2022]
Abstract
AIM Gemcitabine is used not only as a second-line, but also as a third-line or higher regimen for taxane/platinum-resistant recurrent ovarian cancer. The purpose of this study was to clarify the response to and toxicity of gemcitabine for recurrent ovarian cancer according to the number of previous chemotherapy regimens. METHODS The subjects were patients with taxane/platinum-resistant recurrent ovarian cancer on gemcitabine treatment at the present hospital between June 2007 and September 2013. We retrospectively reviewed the medical records. Response and adverse events were assessed using the Response Evaluation Criteria in Solid Tumors version 1.1. and the Common Terminology Criteria for Adverse Events v4.0, respectively. RESULTS The subjects consisted of 65 patients. The median number of previous chemotherapy regimens was 3 (range, 1-7). Overall response rate was 4.6%, and disease control rate (DCR) was 40.0%. DCR versus one, two, three, and ≥four previous chemotherapy regimens was 83.3%, 45.0%, 36.4%, and 23.5%, respectively. Grade 3/4 neutropenia, anemia, and thrombocytopenia occurred in 52.3%, 9.2%, and 9.2% of patients, respectively. Prevalence of grade 3/4 neutropenia according to one, two, three, and ≥four previous chemotherapy regimens was 66.7%, 55.0%, 54.5%, and 41.2%, respectively. Prevalence of anemia, thrombocytopenia, and almost all the non-hematological toxicities also did not increase with an increase in the number of previous chemotherapy regimens. CONCLUSIONS Although DCR decreased as the number of previous chemotherapy regimens increased, the toxicities did not increase. Gemcitabine may be relatively safe in heavily pretreated ovarian cancer patients.
Collapse
Affiliation(s)
- Yuji Takei
- Department of Obstetrics and Gynecology, Jichi Medical University, Tochigi, Japan
| | - Yoshifumi Takahashi
- Department of Obstetrics and Gynecology, Jichi Medical University, Tochigi, Japan
| | - Shizuo Machida
- Department of Obstetrics and Gynecology, Jichi Medical University, Tochigi, Japan
| | - Akiyo Taneichi
- Department of Obstetrics and Gynecology, Jichi Medical University, Tochigi, Japan
| | - Suzuyo Takahashi
- Department of Obstetrics and Gynecology, Jichi Medical University, Tochigi, Japan
| | - Tomomi Nagashima
- Department of Obstetrics and Gynecology, Jichi Medical University, Tochigi, Japan
| | - Hiroyuki Morisawa
- Department of Obstetrics and Gynecology, Jichi Medical University, Tochigi, Japan
| | - Yasushi Saga
- Department of Obstetrics and Gynecology, Jichi Medical University, Tochigi, Japan
| | - Shigeki Matsubara
- Department of Obstetrics and Gynecology, Jichi Medical University, Tochigi, Japan
| | - Hiroyuki Fujiwara
- Department of Obstetrics and Gynecology, Jichi Medical University, Tochigi, Japan
| |
Collapse
|