1
|
Han C, Su J, Pei Y, Su X, Zheng D. LINC00665 promotes the progression and immune evasion of lung cancer by facilitating the translation of TCF7 protein through dependence on IRES. Cancer Cell Int 2024; 24:227. [PMID: 38951802 PMCID: PMC11218341 DOI: 10.1186/s12935-024-03411-4] [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: 02/12/2024] [Accepted: 06/22/2024] [Indexed: 07/03/2024] Open
Abstract
OBJECTIVE To investigate the influence of LINC00665 on the development and immune evasion of lung cancer. METHODS Tumor tissues and corresponding adjacent tissues were collected from 84 lung cancer patients, categorized into non-metastatic (n = 58) and metastatic (n = 26) groups. LINC00665 expression in lung cancer and metastatic lung cancer tissues was assessed via qRT-PCR. Pearson correlation analysis was conducted to examine the correlation between LINC00665 and immune-modulating cytokines (TGF-β, IL-10, IL-1β, IFN-γ, IL-2, TNF-α). A549 and H1299 cells, with relatively high LINC00665 expression, were used for in vitro studies. Cells were transfected with LINC00665-targeting shRNA, and changes in proliferation, apoptosis, migration, invasion, and NK cell cytotoxicity were assessed. Downstream molecular mechanisms of LINC00665 were investigated using GEO database analysis, highlighting the association with HHLA2. LINC00665's role in promoting HHLA2 expression via binding with TCF7 was explored. In low LINC00665-expressing A549/H1299 cells, overexpression of HHLA2 was performed to evaluate effects on malignant behavior and NK cell sensitivity. A xenograft model was established for in vivo validation through tumor volume and weight measurements, Ki-67 immunoreactivity analysis, and flow cytometry analysis of CD107a + NK cells. RESULTS LINC00665, TCF7 mRNA, and HHLA2 mRNA expression levels were significantly higher in lung cancer tissues than adjacent tissues, with non-metastatic lung cancer showing higher expression than metastatic lung cancer. In metastatic lung cancer, LINC00665 positively correlated with immune-suppressive cytokines (TGF-β, IL-10, IL-1β) and negatively correlated with anti-tumor cytokines (IFN-γ, IL-2, TNF-α). LINC00665 knockdown significantly inhibited lung cancer cell growth and metastasis, promoting sensitivity to NK cells. Further analysis revealed that LINC00665 recruits transcription factor TCF7 to upregulate HHLA2 expression in lung cancer cells, thereby facilitating lung cancer development and immune escape. CONCLUSION LINC00665, through recruitment of TCF7 and upregulation of HHLA2, inhibits NK cell cytotoxicity, promoting the development and immune evasion of lung cancer.
Collapse
Affiliation(s)
- Chaonan Han
- Department of Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200443, China
| | - Jinchen Su
- School of Medicine, Shihezi University, Shihezi, 832000, Xinjiang, China
| | - Yue Pei
- Department of Laboratory Medicine, Yixing Hospital of Traditional Chinese Medicine, No.128 East Yangquan Road, Yicheng Subdistrict, Yixing, 214200, Jiangsu, China
| | - Xiangyu Su
- Department of Oncology, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, Jiangsu, P.R. China
| | - Di Zheng
- Department of Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200443, China.
| |
Collapse
|
2
|
Bruno R, Poma AM, Panozzi M, Lenzini A, Elia G, Zirafa CC, Aprile V, Ambrogi MC, Baldini E, Lucchi M, Melfi F, Chella A, Sbrana A, Alì G. Early-Stage Non-Small Cell Lung Cancer: Prevalence of Actionable Alterations in a Monocentric Consecutive Cohort. Cancers (Basel) 2024; 16:1410. [PMID: 38611088 PMCID: PMC11010971 DOI: 10.3390/cancers16071410] [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: 02/20/2024] [Revised: 03/26/2024] [Accepted: 03/31/2024] [Indexed: 04/14/2024] Open
Abstract
Early-stage (ES) non-small cell lung cancer (NSCLC) is diagnosed in about 30% of cases. The preferred treatment is surgery, but a significant proportion of patients experience recurrence. Neoadjuvant and adjuvant chemotherapy has a limited clinical benefit. EGFR tyrosine kinase inhibitors and immunotherapy have recently opened new therapeutic scenarios. However, only a few data are available about the ES-NSCLC molecular landscape and the impact of oncogene addiction on therapy definition. Here, we determined the prevalence of the main lung cancer driver alterations in a monocentric consecutive cohort. Molecular analysis was performed on 1122 cases, including 368 ES and 754 advanced NSCLC. The prevalence of actionable alterations was similar between early and advanced stages. ES-NSCLC was significantly enriched for MET exon-14 skipping alterations and presented a lower prevalence of BRAF p.(V600E) mutation. PD-L1 expression levels, evaluated according to actionable alterations, were higher in advanced than early tumors harboring EGFR, KRAS, MET alterations and gene fusions. Taken together, these results confirm the value of biomarker testing in ES-NSCLC. Although approved targeted therapies for ES-NSCLC are still limited, the identification of actionable alterations could improve patients' selection for immunotherapy, favoring the enrollment in clinical trials and allowing a faster treatment start at disease recurrence.
Collapse
Affiliation(s)
- Rossella Bruno
- Unit of Pathological Anatomy, University Hospital of Pisa, Via Roma 67, 56126 Pisa, Italy;
| | - Anello Marcello Poma
- Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, Via Savi 10, 56126 Pisa, Italy; (A.M.P.); (A.L.); (G.E.); (V.A.); (M.C.A.); (M.L.); (G.A.)
| | - Martina Panozzi
- Unit of Pathological Anatomy, University Hospital of Pisa, Via Roma 67, 56126 Pisa, Italy;
| | - Alessandra Lenzini
- Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, Via Savi 10, 56126 Pisa, Italy; (A.M.P.); (A.L.); (G.E.); (V.A.); (M.C.A.); (M.L.); (G.A.)
| | - Gianmarco Elia
- Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, Via Savi 10, 56126 Pisa, Italy; (A.M.P.); (A.L.); (G.E.); (V.A.); (M.C.A.); (M.L.); (G.A.)
| | - Carmelina Cristina Zirafa
- Minimally Invasive and Robotic Thoracic Surgery, Department of Surgical, Medical, Molecular and Critical Care Pathology, University Hospital of Pisa, Via Paradisa 2, 56124 Pisa, Italy; (C.C.Z.); (F.M.)
| | - Vittorio Aprile
- Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, Via Savi 10, 56126 Pisa, Italy; (A.M.P.); (A.L.); (G.E.); (V.A.); (M.C.A.); (M.L.); (G.A.)
| | - Marcello Carlo Ambrogi
- Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, Via Savi 10, 56126 Pisa, Italy; (A.M.P.); (A.L.); (G.E.); (V.A.); (M.C.A.); (M.L.); (G.A.)
| | - Editta Baldini
- Medical Oncology, Hospital of Lucca, 55100 Lucca, Italy;
| | - Marco Lucchi
- Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, Via Savi 10, 56126 Pisa, Italy; (A.M.P.); (A.L.); (G.E.); (V.A.); (M.C.A.); (M.L.); (G.A.)
| | - Franca Melfi
- Minimally Invasive and Robotic Thoracic Surgery, Department of Surgical, Medical, Molecular and Critical Care Pathology, University Hospital of Pisa, Via Paradisa 2, 56124 Pisa, Italy; (C.C.Z.); (F.M.)
| | - Antonio Chella
- Unit of Pneumology, University Hospital of Pisa, Via Roma 67, 56126 Pisa, Italy; (A.C.); (A.S.)
| | - Andrea Sbrana
- Unit of Pneumology, University Hospital of Pisa, Via Roma 67, 56126 Pisa, Italy; (A.C.); (A.S.)
| | - Greta Alì
- Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, Via Savi 10, 56126 Pisa, Italy; (A.M.P.); (A.L.); (G.E.); (V.A.); (M.C.A.); (M.L.); (G.A.)
| |
Collapse
|
3
|
Zhao ZR, Lin ZC, Shen JF, Xie ZH, Jiang L. Neoadjuvant Immunotherapy in Oncogene-Positive Non-Small Cell Lung Cancer: A Multicenter Study. Ann Thorac Surg 2023; 116:703-710. [PMID: 36521526 DOI: 10.1016/j.athoracsur.2022.11.035] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 11/14/2022] [Accepted: 11/21/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND Preoperative immunotherapy has shed light on the management of resectable non-small cell lung cancer (NSCLC). However, whether neoadjuvant immunotherapy benefits patients with oncogene-positive NSCLC remains unknown. METHODS Data were retrieved from 4 institutions in the period from August 2018 to May 2021. Eligible patients were aged ≥18 years with histologically confirmed stage IIA to stage IIIB (T1-2 N1-2 or T3-4 N0-2) NSCLC that was deemed to be surgically resectable. The neoadjuvant regimen included immune checkpoint inhibitors alone or in combination with platinum-based doublets. Surgical resection was performed 4 to 6 weeks after the first day of the last cycle of treatment. The primary end point was major pathologic response (MPR; ≤10% viable tumor cells). Analyses were categorized according to the patients' oncogene (EGFR, ALK, KRAS, MET, BRAF, ROS1, RET) status. RESULTS Overall, 137 patients were identified; 46 (33%) patients had nonsquamous cell cancer, and 114 (83%) had stage IIIA/B disease. Oncogene alterations were identified in 22 (16%) patients, of whom only 2 patients (2/22 [9%]) had an MPR compared with 65 (65/115 [56.5%]) in the oncogene-negative population (P < .001). Similar results were retained after propensity score matching for age, sex, smoking status, histologic type, stage, and cycles of neoadjuvant treatment. Squamous cell carcinoma (odds ratio, 2.54; 95% CI, 1.08-5.99) and positive oncogene status (odds ratio, 0.13; 95% CI, 0.03-0.64) were found to be indicators for MPR by logistic regression. The 1-year event-free survival rate was 75.4% in the oncogene-positive group, which was not significantly different from 85.5% in the oncogene-negative population (P = .23). CONCLUSIONS Patients with stage II-III oncogene-positive NSCLCs respond less than patients with oncogene-negative NSCLCs after neoadjuvant immunotherapy.
Collapse
Affiliation(s)
- Ze-Rui Zhao
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, and Department of Thoracic Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Zhi-Chao Lin
- Department of Thoracic Surgery, Jiangmen Central Hospital, Jiangmen, China
| | - Jian-Fei Shen
- Department of Cardiothoracic Surgery, Taizhou Hospital, Linhai, China
| | - Ze-Hua Xie
- Department of Thoracic Surgery, Jiangmen Central Hospital, Jiangmen, China
| | - Long Jiang
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
4
|
Bashraheel SS, Goda SK. Novel SPEA Superantigen Peptide Agonists and Peptide Agonist-TGFαL3 Conjugate. In Vitro Study of Their Growth-Inhibitory Effects for Targeted Cancer Immunotherapy. Int J Mol Sci 2023; 24:10507. [PMID: 37445686 DOI: 10.3390/ijms241310507] [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: 05/16/2023] [Revised: 06/13/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023] Open
Abstract
Bacterial superantigens (SAgs) are effective T-cell stimulatory molecules that lead to massive cytokine production. Superantigens crosslink between MHC class II molecules on the Antigen Presenting Cells (APC) and TCR on T-cells. This enables them to activate up to 20% of resting T cells, whilst conventional antigen presentation results in the activation of 0.001-0.0001% of the T cell population. These biological properties of superantigens make them attractive for use in immunotherapy. Previous studies have established the effectiveness of superantigens as therapeutic agents. This, however, was achieved with severe side effects due to the high lethality of the native toxins. Our study aims to produce superantigen-based peptides with minimum or no lethality for safer cancer treatment. In previous work, we designed and synthesized twenty overlapping SPEA-based peptides and successfully mapped regions in SPEA superantigen, causing a vasodilatory response. We screened 20 overlapping SPEA-based peptides designed and synthesized to cover the whole SPEA molecule for T-cell activation and tumor-killing ability. In addition, we designed and synthesized tumor-targeted superantigen-based peptides by fusion of TGFαL3 either from the N' or C' terminal of selected SPEA-based peptides with an eight-amino acid flexible linker in between. Our study identified parts of SPEA capable of stimulating human T-cells and producing different cytokines. We also demonstrated that the SPEA-based peptide conjugate binds specifically to cancer cells and can kill this cancer. Peptides induce T-cell activation, and tumor killing might pave the way for safer tumor-targeted superantigens (TTS). We proposed the combination of our new superantigen-based peptide conjugates with other immunotherapy techniques for effective and safer cancer treatment.
Collapse
Affiliation(s)
| | - Sayed K Goda
- College of Science and Technology, University of Derby, Derby DE22 1GB, UK
| |
Collapse
|
5
|
Shen Q, Xu Z, Sun G, Wang H, Zhang L. TFAP4 Activates IGF2BP1 and Promotes Progression of Non-Small Cell Lung Cancer by Stabilizing TK1 Expression through m6A Modification. Mol Cancer Res 2022; 20:1763-1775. [PMID: 36074102 DOI: 10.1158/1541-7786.mcr-22-0231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 07/12/2022] [Accepted: 09/02/2022] [Indexed: 01/15/2023]
Abstract
Non-small cell lung cancer (NSCLC) is a well-known global health concern. TFAP4 has been reported to function as an oncogene. This study sought to investigate the molecular mechanism of TFAP4 in NSCLC development. Significantly highly-expressed gene IGF2BP1 was screened on online databases and its downstream gene TK1 was predicted. IGF2BP1 promoter sequence was identified. The binding site of TFAP4 and IGF2BP1 was predicted. The expression correlations among TFAP4, IGF2BP1, and TK1 were confirmed. The correlations between TFAP4, IGF2BP1, TK1, and NSCLC prognosis were predicted. NSCLC and paracancerous tissues were collected. The expressions of TFAP4, IGF2BP1, and TK1 were detected. NSCLC cell proliferation, migration, invasion, and apoptosis were detected. The binding of TFAP4 to the IGF2BP1 promoter was verified. m6A modification of TK1 mRNA was detected. The correlation between IGF2BP1 and TK1 was confirmed. A subcutaneous tumor xenograft model was established to validate the effect of TFAP4 in vivo. IGF2BP1 was highly expressed in NSCLC tissues and cells. IGF2BP1 knockdown repressed NSCLC cell proliferation, migration, and invasion and facilitated apoptosis. Mechanically, TFAP4 transcriptionally activated IGF2BP1. IGF2BP1 stabilized TK1 expression via m6A modification and promoted NSCLC cell proliferation, migration, and invasion. In vivo experiments confirmed that TFAP4 knockdown suppressed tumor growth by downregulating IGF2BP1/TK1. IMPLICATIONS Our findings revealed that TFAP4 activated IGF2BP1 and facilitated NSCLC progression by stabilizing TK1 expression via m6A modification, which offered new insights into the diagnosis and treatment of NSCLC.
Collapse
Affiliation(s)
- Qiming Shen
- Department of Thoracic Surgery, The First Hospital of China Medical University, Heping Area, Shenyang, Liaoning, China
| | - Zhe Xu
- Department of Thoracic Surgery, The First Hospital of China Medical University, Heping Area, Shenyang, Liaoning, China
| | - Guanghao Sun
- Department of Thoracic Surgery, The First Hospital of China Medical University, Heping Area, Shenyang, Liaoning, China
| | - Haoyou Wang
- Department of Thoracic Surgery, The First Hospital of China Medical University, Heping Area, Shenyang, Liaoning, China
| | - Lin Zhang
- Department of Thoracic Surgery, The First Hospital of China Medical University, Heping Area, Shenyang, Liaoning, China
| |
Collapse
|
6
|
Rocco D, Della Gravara L, Maione P, Palazzolo G, Gridelli C. Identification of drug combinations for lung cancer patients whose tumors are unresponsive to targeted therapy: clinical bases and future directions. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2022. [DOI: 10.1080/23808993.2022.2050369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Danilo Rocco
- Department of Pulmonary Oncology, AORN dei Colli Monaldi, Naples, Italy
| | - Luigi Della Gravara
- Department of Experimental Medicine, Università degli studi della Campania “Luigi Vanvitelli”, Naples, Italy
| | - Paolo Maione
- Division of Medical Oncology, “S.g. Moscati” Hospital, Avellino, Italy
| | | | - Cesare Gridelli
- Division of Medical Oncology, “S.g. Moscati” Hospital, Avellino, Italy
| |
Collapse
|
7
|
Thomas QD, Basse C, Luporsi M, Girard N. Pembrolizumab Plus Chemotherapy in Metastatic Thymic Carcinoma: A Case Report. Front Oncol 2022; 11:814544. [PMID: 35127529 PMCID: PMC8810527 DOI: 10.3389/fonc.2021.814544] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 12/20/2021] [Indexed: 11/26/2022] Open
Abstract
Metastatic thymic carcinomas have a poor prognosis. Pembrolizumab, an anti-PD-1 antibody, has recently been evaluated for patients with metastatic thymic carcinomas progressing after at least one line of platinum-based chemotherapy. The antitumor activity of immunotherapy appears to be promising for these patients and pembrolizumab in monotherapy is actually a treatment option in second metastatic line. To the best of our knowledge, we report the first case of a patient treated for metastatic thymic adenocarcinoma with a combination of chemotherapy–immunotherapy. The patient is a 46-year-old man with metastatic thymic adenocarcinoma treated in third metastatic line with a combination of pembrolizumab plus platinum-based chemotherapy with a very good metabolic tumor response. He had a progression-free survival of 7.9 months and did not experience any severe side effects related to pembrolizumab. The association of immunotherapy and chemotherapy, as in non-small cell and small cell lung cancers, could be of interest for future therapeutic trials evaluating the survival of patients with metastatic thymic carcinoma.
Collapse
Affiliation(s)
- Quentin Dominique Thomas
- Department of Medical Oncology, Montpellier Cancer institute (ICM), Montpellier, France
- Oncogenic Pathways in Lung Cancer, Montpellier Cancer Research Institute (IRCM), University of Montpellier (UM), Montpellier, France
- *Correspondence: Quentin Dominique Thomas,
| | - Clémence Basse
- Department of Medical Oncology, Curie Montsouris Thorax Institute, Institut Curie, Paris, France
| | - Marie Luporsi
- Department of Nuclear Medicine, Institut Curie, Paris, France
| | - Nicolas Girard
- Department of Medical Oncology, Curie Montsouris Thorax Institute, Institut Curie, Paris, France
| |
Collapse
|
8
|
Martin C, Cuello M, Barajas O, Recondo G, Aruachan S, Perroud H, Sena S, Bonilla C, Orlandi F, Berutti S, Garcia Cocco V, Gomez A, Korbenfeld E, Zapata M, Cundom J, Orellana E, Goncalves S, Reinhold F. Real-world evaluation of molecular testing and treatment patterns for EGFR mutations in non-small cell lung cancer in Latin America. Mol Clin Oncol 2021; 16:6. [PMID: 34881026 PMCID: PMC8647188 DOI: 10.3892/mco.2021.2439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 09/08/2021] [Indexed: 12/24/2022] Open
Abstract
Lung cancer is a leading cause of cancer-related deaths in Latin America, with non-small cell lung cancer (NSCLC) being the most prevalent. The current study aimed to report real-world data on epidermal growth factor receptor (EGFR) mutational testing and treatment regimens at diagnosis and progression in patients with metastatic NSCLC across four Latin American countries (Argentina, Chile, Colombia and Uruguay). A retrospective, multicenter, observational study was conducted in patients with NSCLC using medical records from participating countries. The study population was categorized into two cohorts: Cohort 1 comprised of newly diagnosed, treatment-naïve patients with stage IV NSCLC; and cohort 2 comprised of stage IV NSCLC EGFR mutation (EGFRm)-positive patients who had progressed after first- or second-generation EGFR-tyrosine kinase inhibitor (TKI) treatment. Measures included demographic variables, health characteristics, treatment regimen, molecular testing rate and turnaround time at diagnosis and at progression for cohorts 1 and 2, respectively. Descriptive statistics were used to summarize all study measures. Of the 462 patients enrolled, 431 were newly diagnosed or treatment naïve with metastatic NSCLC. In cohort 1, the majority of patients with private health insurance (57.31%) underwent molecular diagnosis while only 41.3% of patients within the public sector had access to testing. The average molecular testing rate in cohort 1 varied across countries, with Argentina having the highest testing rate (79%) and Uruguay the lowest (27.63%). EGFRm was observed in 22% of patients. Cohort 2 comprised 31 patients who had progressed after first- or second-generation EGFR-TKI treatment and of these, only 22 (70.97%) underwent testing after progression. Access to molecular testing is still a challenge impacting the choice of first-line treatment in Latin American patients with NSCLC. These findings underline the unmet needs of ensuring early diagnosis, molecular profiling and use of correct treatment to alleviate NSCLC burden in the region.
Collapse
Affiliation(s)
- Claudio Martin
- Department of Clinical Oncology, Institute Alexander Fleming, Buenos Aires C1426ANZ, Argentina
| | - Mauricio Cuello
- Department of Oncology, Hospital of Clinics, Montevideo 11600, Uruguay
| | - Olga Barajas
- Department of Medical Oncology, Arturo Lopez Perez Foundation, Santiago 7500000, Chile
| | - Gonzalo Recondo
- Department of Thoracic Oncology, Medical education and Clinical Research Centre, University Institute (CEMIC), Buenos Aires C1431FWO, Argentina
| | - Sandra Aruachan
- Department of Clinical Oncology, High Technology Medical Institute Oncomedica, Monteria 23001, Colombia
| | - Herman Perroud
- Department of Clinical Oncology, Women's Health Centre, Rosario 2000, Argentina
| | - Susana Sena
- Department of Clinical Oncology; German Hospital, Buenos Aires PC C1118AAT, Argentina
| | - Carlos Bonilla
- Clinical Oncology Unit, National Cancer Institute of Colombia, Bogotá 110411, Colombia
| | - Francisco Orlandi
- Department of Clinical Oncology, Orlandi Oncology, Providencia 7500713, Chile
| | - Susana Berutti
- Department of Clinical Oncology, Italian Hospital of La Plata, La Plata B1900AXI, Argentina
| | | | - Alvaro Gomez
- Department of Medical Oncology, Hemato-oncologos SA, Cali 760042, Colombia
| | - Ernesto Korbenfeld
- Department of Medical Oncology, British Hospital of Buenos Aires, Buenos Aires C1280AEB, Argentina
| | - Maycos Zapata
- Department of Medical Oncology, Cancer Institute Las Americas, Antioquia 050025, Colombia
| | - Juan Cundom
- Department of Medical Oncology, Lanari Institute, University of Buenos Aires, Buenos Aires C1427ARN, Argentina
| | - Eric Orellana
- Department of Clinical Oncology, Clinic Santa Maria, Santiago 7500000, Chile
| | | | | |
Collapse
|
9
|
Comparison of the outcome between immunotherapy alone or in combination with chemotherapy in EGFR-mutant non-small cell lung cancer. Sci Rep 2021; 11:16122. [PMID: 34373555 PMCID: PMC8352947 DOI: 10.1038/s41598-021-95628-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 07/22/2021] [Indexed: 12/17/2022] Open
Abstract
Whether ICIs combined with chemotherapy can improve outcomes in EGFR-mutant non-small cell lung cancer (NSCLC) remains uncertain. Patients with EGFR-mutant NSCLC and who progressed on first-line EGFR-TKIs treatment were retrospectively collected. We reviewed the outcome of these patients treated with ICIs or ICIs combined chemotherapy (ICI + C). Total 30 patients were included. The ORR were 9.1% and 25.0% for the ICI and ICI + C groups. The ICI + C group showed the trend of longer progression-free survival and overall survival periods. Patients without the T790M mutation had a significantly longer PFS than did those without this mutation (4.23 [95% CI: 2.75–5.72] vs. 1.70 [95% CI: 0.00–3.51] months, HR:4.45, p = 0.019). ICIs combined with chemotherapy tended to be more effective than ICIs alone in pretreated EGFR-mutant NSCLC. The T790M mutation may be a potential biomarker.
Collapse
|
10
|
Poma AM, Bruno R, Pietrini I, Alì G, Pasquini G, Proietti A, Vasile E, Cappelli S, Chella A, Fontanini G. Biomarkers and Gene Signatures to Predict Durable Response to Pembrolizumab in Non-Small Cell Lung Cancer. Cancers (Basel) 2021; 13:cancers13153828. [PMID: 34359727 PMCID: PMC8345106 DOI: 10.3390/cancers13153828] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/27/2021] [Accepted: 07/28/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Not all patients with advanced or metastatic non-small cell lung cancer (NSCLC) respond to pembrolizumab, even if their tumor expresses PD-L1. This is a monocentric study aimed at identifying potential predictive biomarkers for pembrolizumab first-line treatment. Tumor microenvironment was characterized by gene expression analysis in 46 tumor samples from 25 NSCLC patients with and 21 without durable clinical benefit. As expected, patients achieving clinical benefit had a greater infiltration of immune cells. In particular, CD8 T-cell and NK cell scores were strongly associated with durable benefit. Single immune cell markers such as XCL1/2 showed a high performance in predicting durable response to pembrolizumab with an AUC of 0.85. In the same series PD-L1 expression levels had an AUC equal to 0.61. Identified predictive biomarkers can improve patients’ selection, thus optimizing treatment definition. Abstract Pembrolizumab has been approved as first-line treatment for advanced Non-small cell lung cancer (NSCLC) patients with tumors expressing PD-L1 and in the absence of other targetable alterations. However, not all patients that meet these criteria have a durable benefit. In this monocentric study, we aimed at refining the selection of patients based on the expression of immune genes. Forty-six consecutive advanced NSCLC patients treated with pembrolizumab in first-line setting were enrolled. The expression levels of 770 genes involved in the regulation of the immune system was analysed by the nanoString system. PD-L1 expression was evaluated by immunohistochemistry. Patients with durable clinical benefit had a greater infiltration of cytotoxic cells, exhausted CD8, B-cells, CD45, T-cells, CD8 T-cells and NK cells. Immune cell scores such as CD8 T-cell and NK cell were good predictors of durable response with an AUC of 0.82. Among the immune cell markers, XCL1/2 showed the better performance in predicting durable benefit to pembrolizumab, with an AUC of 0.85. Additionally, CD8A, CD8B and EOMES showed a high specificity (>0.86) in identifying patients with a good response to treatment. In the same series, PD-L1 expression levels had an AUC of 0.61. The characterization of tumor microenvironment, even with the use of single markers, can improve patients’ selection for pembrolizumab treatment.
Collapse
Affiliation(s)
- Anello Marcello Poma
- Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, via Savi 10, 56126 Pisa, Italy;
| | - Rossella Bruno
- Unit of Pathological Anatomy, University Hospital of Pisa, via Roma 67, 56126 Pisa, Italy; (R.B.); (G.A.); (A.P.)
| | - Iacopo Pietrini
- General Pathology, University of Pisa, via Savi 10, 56126 Pisa, Italy;
| | - Greta Alì
- Unit of Pathological Anatomy, University Hospital of Pisa, via Roma 67, 56126 Pisa, Italy; (R.B.); (G.A.); (A.P.)
| | - Giulia Pasquini
- Unit of Medical Oncology, San Jacopo Hospital of Pistoia, 51100 Pistoia, Italy;
| | - Agnese Proietti
- Unit of Pathological Anatomy, University Hospital of Pisa, via Roma 67, 56126 Pisa, Italy; (R.B.); (G.A.); (A.P.)
| | - Enrico Vasile
- Unit of Pneumology, University Hospital of Pisa, via Paradisa 2, 56126 Pisa, Italy; (E.V.); (S.C.); (A.C.)
| | - Sabrina Cappelli
- Unit of Pneumology, University Hospital of Pisa, via Paradisa 2, 56126 Pisa, Italy; (E.V.); (S.C.); (A.C.)
| | - Antonio Chella
- Unit of Pneumology, University Hospital of Pisa, via Paradisa 2, 56126 Pisa, Italy; (E.V.); (S.C.); (A.C.)
| | - Gabriella Fontanini
- Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, via Savi 10, 56126 Pisa, Italy;
- Correspondence: ; Tel.: +39-050-992983
| |
Collapse
|
11
|
Emerging Biomarkers for the Selection of Advanced NSCLC-Affected Immunotherapy Patients. JOURNAL OF MOLECULAR PATHOLOGY 2021. [DOI: 10.3390/jmp2020017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Immunotherapy in the form of ICIs has revolutionized advanced NSCLC treatment algorithms, with ICI-containing combination treatments being the latest addition to approved regimens. However, PD-L1 still represents the only routinely assessed and validated biomarker apart from genetic drivers testing, impairing our capacity to personalize and guide treatment. Therefore, this paper aims to analyze the most promising emerging predictive biomarkers that could help us in the near future to select patients more effectively.
Collapse
|
12
|
El Husseini K, Wislez M. Sequential or combined immune checkpoint inhibitors and targeted therapy: Navigating uncharted waters. Respir Med Res 2021; 79:100820. [PMID: 33892315 DOI: 10.1016/j.resmer.2021.100820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/26/2021] [Accepted: 03/27/2021] [Indexed: 10/21/2022]
Affiliation(s)
- K El Husseini
- Thoracic Oncology Unit, Department of Pneumology, HUPC, groupe hospitalier, hôpital Cochin, AP-HP, 27, rue du Faubourg Saint-Jacques, Paris, France
| | - M Wislez
- Thoracic Oncology Unit, Department of Pneumology, HUPC, groupe hospitalier, hôpital Cochin, AP-HP, 27, rue du Faubourg Saint-Jacques, Paris, France.
| |
Collapse
|
13
|
Yamaguchi M, Nakagawa K, Suzuki K, Takamochi K, Ito H, Okami J, Aokage K, Shiono S, Yoshioka H, Aoki T, Tsutani Y, Okada M, Watanabe SI. Surgical challenges in multimodal treatment of N2-stage IIIA non-small cell lung cancer. Jpn J Clin Oncol 2021; 51:333-344. [PMID: 33506253 DOI: 10.1093/jjco/hyaa249] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 11/24/2020] [Indexed: 12/25/2022] Open
Abstract
Locally advanced non-small cell lung cancer, especially mediastinal lymph node metastasis-positive stage IIIA-N2 cancer, is a heterogeneous disease state characterized by anatomically locally advanced disease with latent micrometastases. Thus, surgical resection or radiotherapy alone has historically failed to cure this disease. During the last three decades, persistent efforts have been made to develop a suitable treatment modality to overcome these problems using chemotherapy and/or radiotherapy with surgical resection. However, the role of surgical resection remains unclear, and the standard treatment for stage IIIA-N2 disease is concurrent chemoradiotherapy. In general, adjuvant chemotherapy is indicated for completely resected pathological stage IB disease or lymph node metastasis-positive pathological stage II or IIIA disease. Platinum-based doublet cytotoxic chemotherapy is currently the standard regimen. Additionally, post-operative radiotherapy might be indicated for post-operatively proven mediastinal lymph node metastasis; i.e. clinical N0-1 and pathological N2 disease. With the remarkable progression that has recently been made in the field of chemotherapy, such as advances in molecular targeting agents and immune checkpoint inhibitors, the basic policy of chemotherapy has been shifting to personalized treatment based on the individual patient's oncogene driver mutation status, immune status and other parameters. The same trend is being seen in the treatment of stage IIIA-N2 disease. We should consider the past and upcoming results of several clinical trials to optimize the coming era of personalized treatment.
Collapse
Affiliation(s)
- Masafumi Yamaguchi
- Department of Thoracic Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan
| | - Kazuo Nakagawa
- Department of Thoracic Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Kenji Suzuki
- Department of General Thoracic Surgery, Juntendo University Hospital, Tokyo, Japan
| | - Kazuya Takamochi
- Department of General Thoracic Surgery, Juntendo University Hospital, Tokyo, Japan
| | - Hiroyuki Ito
- Department of Thoracic Surgery, Kanagawa Cancer Center, Kanagawa, Japan
| | - Jiro Okami
- Department of General Thoracic Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Keiju Aokage
- Department of Thoracic Surgery, National Cancer Center Hospital East, Chiba, Japan
| | - Satoshi Shiono
- Department of Thoracic Surgery, Yamagata Prefectural Central Hospital, Yamagata, Japan
| | - Hiroshige Yoshioka
- Department of Thoracic Oncology, Kansai Medical University Hospital, Osaka, Japan
| | - Tadashi Aoki
- Department of Thoracic Surgery, Niigata Cancer Center Hospital, Niigata, Japan
| | - Yasuhiro Tsutani
- Department of Surgical Oncology, Hiroshima University, Hiroshima, Japan
| | - Morihito Okada
- Department of Surgical Oncology, Hiroshima University, Hiroshima, Japan
| | - Shun-Ichi Watanabe
- Department of Thoracic Surgery, National Cancer Center Hospital, Tokyo, Japan
| | | |
Collapse
|
14
|
Chen YJ, Roumeliotis TI, Chang YH, Chen CT, Han CL, Lin MH, Chen HW, Chang GC, Chang YL, Wu CT, Lin MW, Hsieh MS, Wang YT, Chen YR, Jonassen I, Ghavidel FZ, Lin ZS, Lin KT, Chen CW, Sheu PY, Hung CT, Huang KC, Yang HC, Lin PY, Yen TC, Lin YW, Wang JH, Raghav L, Lin CY, Chen YS, Wu PS, Lai CT, Weng SH, Su KY, Chang WH, Tsai PY, Robles AI, Rodriguez H, Hsiao YJ, Chang WH, Sung TY, Chen JS, Yu SL, Choudhary JS, Chen HY, Yang PC, Chen YJ. Proteogenomics of Non-smoking Lung Cancer in East Asia Delineates Molecular Signatures of Pathogenesis and Progression. Cell 2021; 182:226-244.e17. [PMID: 32649875 DOI: 10.1016/j.cell.2020.06.012] [Citation(s) in RCA: 221] [Impact Index Per Article: 55.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 03/13/2020] [Accepted: 06/03/2020] [Indexed: 12/13/2022]
Abstract
Lung cancer in East Asia is characterized by a high percentage of never-smokers, early onset and predominant EGFR mutations. To illuminate the molecular phenotype of this demographically distinct disease, we performed a deep comprehensive proteogenomic study on a prospectively collected cohort in Taiwan, representing early stage, predominantly female, non-smoking lung adenocarcinoma. Integrated genomic, proteomic, and phosphoproteomic analysis delineated the demographically distinct molecular attributes and hallmarks of tumor progression. Mutational signature analysis revealed age- and gender-related mutagenesis mechanisms, characterized by high prevalence of APOBEC mutational signature in younger females and over-representation of environmental carcinogen-like mutational signatures in older females. A proteomics-informed classification distinguished the clinical characteristics of early stage patients with EGFR mutations. Furthermore, integrated protein network analysis revealed the cellular remodeling underpinning clinical trajectories and nominated candidate biomarkers for patient stratification and therapeutic intervention. This multi-omic molecular architecture may help develop strategies for management of early stage never-smoker lung adenocarcinoma.
Collapse
Affiliation(s)
- Yi-Ju Chen
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan
| | - Theodoros I Roumeliotis
- Functional Proteomics Group, Chester Beatty Laboratories, The Institute of Cancer Research, London SW3 6JB, UK
| | - Ya-Hsuan Chang
- Institute of Statistical Science, Academia Sinica, Taipei, Taiwan
| | - Ching-Tai Chen
- Institute of Information Science, Academia Sinica, Taipei, Taiwan
| | - Chia-Li Han
- Master Program in Clinical Pharmacogenomics and Pharmacoproteomics, College of Pharmacy, Taipei Medical University, Taipei, Taiwan.
| | - Miao-Hsia Lin
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan
| | - Huei-Wen Chen
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Gee-Chen Chang
- Division of Chest Medicine, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan; Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Yih-Leong Chang
- Department of Pathology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chen-Tu Wu
- Department of Pathology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Mong-Wei Lin
- Division of Thoracic Surgery, Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Min-Shu Hsieh
- Department of Pathology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yu-Tai Wang
- National Applied Research Laboratories, National Center for High-performance Computing, Hsinchu, Taiwan
| | - Yet-Ran Chen
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei, Taiwan
| | - Inge Jonassen
- Computational Biology Unit (CBU), Informatics Department, University of Bergen, Bergen, Norway
| | | | - Ze-Shiang Lin
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Kuen-Tyng Lin
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan
| | - Ching-Wen Chen
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Pei-Yuan Sheu
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chen-Ting Hung
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | | | - Hao-Chin Yang
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan
| | - Pei-Yi Lin
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan
| | - Ta-Chi Yen
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan
| | - Yi-Wei Lin
- Division of Thoracic Surgery, Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Jen-Hung Wang
- Institute of Information Science, Academia Sinica, Taipei, Taiwan
| | - Lovely Raghav
- Institute of Statistical Science, Academia Sinica, Taipei, Taiwan; Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Bioinformatics Program, Taiwan International Graduate Program, Hsinchu, Taiwan
| | - Chien-Yu Lin
- Institute of Statistical Science, Academia Sinica, Taipei, Taiwan
| | - Yan-Si Chen
- Institute of Statistical Science, Academia Sinica, Taipei, Taiwan
| | - Pei-Shan Wu
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan
| | - Chi-Ting Lai
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan
| | | | - Kang-Yi Su
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Wei-Hung Chang
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei, Taiwan
| | - Pang-Yan Tsai
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei, Taiwan
| | - Ana I Robles
- Office of Cancer Clinical Proteomics Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Henry Rodriguez
- Office of Cancer Clinical Proteomics Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yi-Jing Hsiao
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Wen-Hsin Chang
- Institute of Molecular Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Ting-Yi Sung
- Institute of Information Science, Academia Sinica, Taipei, Taiwan.
| | - Jin-Shing Chen
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan.
| | - Sung-Liang Yu
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan.
| | - Jyoti S Choudhary
- Functional Proteomics Group, Chester Beatty Laboratories, The Institute of Cancer Research, London SW3 6JB, UK.
| | - Hsuan-Yu Chen
- Institute of Statistical Science, Academia Sinica, Taipei, Taiwan; Ph.D. Program in Microbial Genomics, National Chung Hsing University, Taichung, Taiwan.
| | - Pan-Chyr Yang
- Department of Internal Medicine, National Taiwan University, Taipei, Taiwan; Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
| | - Yu-Ju Chen
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan; Department of Chemistry, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
15
|
Zhang M, Rodrigues AJ, Pollom EL, Gibbs IC, Soltys SG, Hancock SL, Neal JW, Padda SK, Ramchandran KJ, Wakelee HA, Chang SD, Lim M, Hayden Gephart M, Li G. Improved survival and disease control following pembrolizumab-induced immune-related adverse events in high PD-L1 expressing non-small cell lung cancer with brain metastases. J Neurooncol 2021; 152:125-134. [PMID: 33415659 DOI: 10.1007/s11060-020-03686-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 12/22/2020] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Immune checkpoint inhibitors have become standard of care for many patients with non-small cell lung cancer (NSCLC). These agents often cause immune-related adverse events (IRAEs), which have been associated with increased overall survival (OS). Intracranial disease control and OS for patients experiencing IRAEs with metastatic NSCLC and brain metastases have not yet been described. METHODS We performed a single-institution, retrospective review of patients with NSCLC and existing diagnosis of brain metastasis, who underwent pembrolizumab treatment and developed any grade IRAE. The primary outcome of the study was intracranial time to treatment failure (TTF), defined from time of pembrolizumab initiation to new intracranial disease progression or death. Kaplan-Meier and Cox proportional hazard analyses were performed. RESULTS A total of 63 patients with NSCLC brain metastasis were identified, and 24 developed IRAEs. Patients with any grade IRAEs had longer OS (21 vs. 10 months, p = 0.004), systemic TTF (15 vs. 4 months, p < 0.001) and intracranial TTF (14 vs. 5 months, p = 0.001), relative to patients without IRAEs. Presence of IRAEs and high PD-L1 (≥ 50%), but not absent/moderate PD-L1 (0-49%), had a positive association for OS, systemic TTF, and intracranial TTF. Following multivariable analysis, IRAE experienced on pembrolizumab was an independent predictor of OS, systemic TTF, and intracranial TTF. CONCLUSIONS In our series of patients with NSCLC and brain metastases treated with pembrolizumab, IRAE presence was associated with a significant increase in OS, systemic TTF, and intracranial TTF. Future studies with increased cohorts will clarify how IRAEs should be interpreted among molecular subtypes.
Collapse
Affiliation(s)
- Michael Zhang
- Department of Neurosurgery, Stanford Medical Center, Palo Alto, CA, 94304, USA
| | - Adrian J Rodrigues
- Department of Neurosurgery, Stanford Medical Center, Palo Alto, CA, 94304, USA
| | - Erqi L Pollom
- Department of Radiation Oncology, Stanford Medical Center, Palo Alto, CA, 94304, USA
| | - Iris C Gibbs
- Department of Radiation Oncology, Stanford Medical Center, Palo Alto, CA, 94304, USA
| | - Scott G Soltys
- Department of Radiation Oncology, Stanford Medical Center, Palo Alto, CA, 94304, USA
| | - Steven L Hancock
- Department of Radiation Oncology, Stanford Medical Center, Palo Alto, CA, 94304, USA
| | - Joel W Neal
- Department of Medicine, Stanford Medical Center, Palo Alto, CA, 94304, USA
| | - Sukhmani K Padda
- Department of Medicine, Stanford Medical Center, Palo Alto, CA, 94304, USA
| | | | - Heather A Wakelee
- Department of Medicine, Stanford Medical Center, Palo Alto, CA, 94304, USA
| | - Steven D Chang
- Department of Neurosurgery, Stanford Medical Center, Palo Alto, CA, 94304, USA
| | - Michael Lim
- Department of Neurosurgery, Stanford Medical Center, Palo Alto, CA, 94304, USA
| | | | - Gordon Li
- Department of Neurosurgery, Stanford Medical Center, Palo Alto, CA, 94304, USA.
| |
Collapse
|
16
|
Lau SCM, Fares AF, Le LW, Mackay KM, Soberano S, Chan SW, Smith E, Ryan M, Tsao MS, Bradbury PA, Pal P, Shepherd FA, Liu G, Leighl NB, Sacher AG. Subtypes of EGFR- and HER2-Mutant Metastatic NSCLC Influence Response to Immune Checkpoint Inhibitors. Clin Lung Cancer 2021; 22:253-259. [PMID: 33582070 DOI: 10.1016/j.cllc.2020.12.015] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 12/18/2020] [Accepted: 12/23/2020] [Indexed: 11/16/2022]
Abstract
INTRODUCTION The efficacy of immune checkpoint inhibitors (ICIs) is low among EGFR-mutated non-small-cell lung cancer (NSCLC), although prolonged responses have occasionally been reported. We investigated the association between mutation subtypes and ICI outcomes among HER2- and EGFR-mutated NSCLC. PATIENTS AND METHODS This retrospective single-center study analyzed patients with EGFR- and HER2-mutated advanced NSCLC who received at least 1 cycle of ICI between 2013 and 2019. Patient characteristics, mutation subtype, and ICI outcomes. RESULTS Among 48 patients with advanced NSCLC, 14 (29%) had HER2 mutations and 34 (71%) had EGFR mutations. EGFR mutations included 16 (47%) exon 19 deletion, 7 (21%) L858R, 5 (15%) uncommon, and 6 (18%) exon 20 insertion. Compared to EGFR-sensitizing mutations (ESMs), HER2 and EGFR exon 20 mutations were associated with a trend toward better response (respectively, ESM, HER2, and EGFR exon 20: 11%, 29%, and 50%; P = .07) and significantly better disease control rates (respectively, 18%, 57%, and 67%; P = .008). Compared to ESM, HER2 mutations (adjusted hazard ratio, 0.35; P = .02) and EGFR exon 20 mutations (adjusted hazard ratio, 0.37; P = .10 trend) were also associated with improved PFS. Programmed death ligand 1 (PD-L1) expression remained an independent predictor of PFS (adjusted hazard ratio, 0.42; 95% confidence interval, 0.23-0.76; P = .004). The 6-month PFS rates were 29% (HER2), 33% (EGFR exon 20), and 4% (ESM). ICIs were generally well tolerated in this population. Importantly, no immune-related toxicity was observed in 10 patients who received a tyrosine kinase inhibitor (TKI) as the immediate next line treatment after ICI. CONCLUSION HER2 and EGFR exon 20 mutations derive greater benefit from ICIs with comparable PFS to wild-type historical second/third-line unselected cohorts. ICIs remain a treatment option for this genomic subgroup, given the absence of approved targeted therapies for these rare mutations.
Collapse
Affiliation(s)
- Sally C M Lau
- Department of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Canada
| | - Aline Fusco Fares
- Department of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Canada
| | - Lisa W Le
- Department of Biostatistics, Princess Margaret Cancer Centre, Toronto, Canada
| | - Kate M Mackay
- Department of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Canada
| | - Spencer Soberano
- Department of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Canada
| | - Sze Wah Chan
- Department of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Canada
| | - Elliot Smith
- Department of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Canada
| | - Malcolm Ryan
- Department of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Canada
| | - Ming Sound Tsao
- Department of Pathology, Laboratory Medicine Program, University Health Network and University of Toronto, Toronto, Canada
| | - Penelope A Bradbury
- Department of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Canada
| | - Prodipto Pal
- Department of Pathology, Laboratory Medicine Program, University Health Network and University of Toronto, Toronto, Canada
| | - Frances A Shepherd
- Department of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Canada
| | - Geoffrey Liu
- Department of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Canada
| | - Natasha B Leighl
- Department of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Canada
| | - Adrian G Sacher
- Department of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Canada; Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, Canada.
| |
Collapse
|
17
|
Kim SR, Chun SH, Kim JR, Kim SY, Seo JY, Jung CK, Gil BM, Kim JO, Ko YH, Woo IS, Shim BY, Hong SH, Kang JH. The implications of clinical risk factors, CAR index, and compositional changes of immune cells on hyperprogressive disease in non-small cell lung cancer patients receiving immunotherapy. BMC Cancer 2021; 21:19. [PMID: 33402127 PMCID: PMC7786505 DOI: 10.1186/s12885-020-07727-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 12/11/2020] [Indexed: 01/19/2023] Open
Abstract
Background Immune checkpoint blockades (ICBs) are characterized by a durable clinical response and better tolerability in patients with a variety of advanced solid tumors. However, we not infrequently encounter patients with hyperprogressive disease (HPD) exhibiting paradoxically accelerated tumor growth with poor clinical outcomes. This study aimed to investigate implications of clinical factors and immune cell composition on different tumor responses to immunotherapy in patients with non-small cell lung cancer (NSCLC). Methods This study evaluated 231 NSCLC patients receiving ICBs between January 2014 and May 2018. HPD was defined as a > 2-fold tumor growth kinetics ratio during ICB therapy and time-to-treatment failure of ≤2 months. We analyzed clinical data, imaging studies, periodic serologic indexes, and immune cell compositions in tumors and stromata using multiplex immunohistochemistry. Results Of 231 NSCLC patients, PR/CR and SD were observed in 50 (21.6%) and 79 (34.2%) patients, respectively and 26 (11.3%) patients met the criteria for HPD. Median overall survival in poor response groups (HPD and non-HPD PD) was extremely shorter than disease-controlled group (SD and PR/CR) (5.5 and 6.1 months vs. 16.2 and 18.3 months, respectively, P = 0.000). In multivariate analysis, HPD were significantly associated with heavy smoker (p = 0.0072), PD-L1 expression ≤1% (p = 0.0355), and number of metastatic site ≥3 (p = 0.0297). Among the serologic indexes including NLR, PLR, CAR, and LDH, only CAR had constantly significant correlations with HPD at the beginning of prior treatment and immunotherapy, and at the 1st tumor assessment. The number of CD4+ effector T cells and CD8+ cytotoxic T cells, and CD8+/PD-1+ tumor-infiltrating lymphocytes (TIL) tended to be smaller, especially in stromata of HPD group. More M2-type macrophages expressing CD14, CD68 and CD163 in the stromal area and markedly fewer CD56+ NK cells in the intratumoral area were observed in HPD group. Conclusions Our study suggests that not only clinical factors including heavy smoker, very low PD-L1 expression, multiple metastasis, and CAR index, but also fewer CD8+/PD-1+ TIL and more M2 macrophages in the tumor microenvironment are significantly associated with the occurrence of HPD in the patients with advanced/metastatic NSCLC receiving immunotherapy. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-020-07727-y.
Collapse
Affiliation(s)
- Seo Ree Kim
- Division of Medical Oncology, Department of Internal Medicine, Bucheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sang Hoon Chun
- Division of Medical Oncology, Department of Internal Medicine, Bucheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Joo Ri Kim
- Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Sang-Yeob Kim
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jun Young Seo
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Chan Kwon Jung
- Department of Hospital Pathology, College of Medicine, the Catholic University of Korea, Seoul, Republic of Korea
| | - Bo-Mi Gil
- Department of Radiology, Bucheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jeong-Oh Kim
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yoon Ho Ko
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Division of Medical Oncology, Department of Internal Medicine, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - In Sook Woo
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Division of Medical Oncology, Department of Internal Medicine, Yeouido St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Byoung Yong Shim
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Division of Medical Oncology, Department of Internal Medicine, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sook-Hee Hong
- Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea.,Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Laboratory of Medical Oncology, Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jin Hyoung Kang
- Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea. .,Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea. .,Laboratory of Medical Oncology, Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| |
Collapse
|
18
|
D'Andréa G, Lassalle S, Guevara N, Mograbi B, Hofman P. From biomarkers to therapeutic targets: the promise of PD-L1 in thyroid autoimmunity and cancer. Theranostics 2021; 11:1310-1325. [PMID: 33391536 PMCID: PMC7738901 DOI: 10.7150/thno.50333] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 08/30/2020] [Indexed: 12/12/2022] Open
Abstract
The programmed cell death-1/programmed cell death ligand-1 (PD-1/PD-L1) immune checkpoint proteins hold promise as diagnostic, prognostic, and therapeutic targets for precision oncology. By restoring antitumor T cell surveillance, the high degree of effectiveness of the immune checkpoint inhibitors (ICIs) has revolutionized cancer treatment. However, the majority of patients (65-80 %) treated with ICIs experience significant side effects, called immune-related adverse events (irAEs), resulting in autoimmune damage to various organs. Therefore, broadening the clinical applicability of these treatments to all cancer types requires an improved understanding of the mechanisms linking cancer immune evasion and autoimmunity. The thyroid is the endocrine gland the most frequently involved in autoimmunity and cancer, the growing incidence of which is raising serious public health issues worldwide. In addition, the risk of developing thyroid cancer is increased in patients with autoimmune thyroid disease and thyroid dysfunction is one of the most common irAEs, especially with PD‑1/PD-L1 blockade. Therefore, we chose the thyroid as a model for the study of the link between autoimmunity, irAEs, and cancer. We provide an update into the current knowledge of the PD‑1/PD-L1 axis and discuss the growing interest of this axis in the diagnosis, prognosis, and management of thyroid diseases within the context of autoimmunity and cancer, while embracing personalized medicine.
Collapse
Affiliation(s)
- Grégoire D'Andréa
- ENT and Head and Neck surgery department, Institut Universitaire de la Face et du Cou, CHU de Nice, University Hospital, Côte d'Azur University, Nice, France
- Côte d'Azur University, CNRS, INSERM, Institute for Research on Cancer and Aging, FHU OncoAge, Nice, France
| | - Sandra Lassalle
- Côte d'Azur University, CNRS, INSERM, Institute for Research on Cancer and Aging, FHU OncoAge, Nice, France
- Laboratory of Clinical and Experimental Pathology, Côte d'Azur University and Biobank, Pasteur Hospital, University Côte d'Azur, FHU OncoAge, Nice, France
| | - Nicolas Guevara
- ENT and Head and Neck surgery department, Institut Universitaire de la Face et du Cou, CHU de Nice, University Hospital, Côte d'Azur University, Nice, France
| | - Baharia Mograbi
- Côte d'Azur University, CNRS, INSERM, Institute for Research on Cancer and Aging, FHU OncoAge, Nice, France
- Antoine Lacassagne Cancer Center, FHU OncoAge, Nice, France
| | - Paul Hofman
- Côte d'Azur University, CNRS, INSERM, Institute for Research on Cancer and Aging, FHU OncoAge, Nice, France
- Antoine Lacassagne Cancer Center, FHU OncoAge, Nice, France
- Laboratory of Clinical and Experimental Pathology, Côte d'Azur University and Biobank, Pasteur Hospital, University Côte d'Azur, FHU OncoAge, Nice, France
| |
Collapse
|
19
|
Imyanitov EN, Iyevleva AG, Levchenko EV. Molecular testing and targeted therapy for non-small cell lung cancer: Current status and perspectives. Crit Rev Oncol Hematol 2020; 157:103194. [PMID: 33316418 DOI: 10.1016/j.critrevonc.2020.103194] [Citation(s) in RCA: 299] [Impact Index Per Article: 59.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 11/14/2020] [Accepted: 11/17/2020] [Indexed: 02/07/2023] Open
Abstract
Molecular testing has become a mandatory component of the non-small cell lung cancer (NSCLC) management. The detection of EGFR, BRAF and MET mutations as well as the analysis of ALK, ROS1, RET and NTRK translocations have already been incorporated in the NSCLC diagnostic standards, and the inhibitors of these kinases are in routine clinical use. There are emerging biomarkers, e.g., KRAS G12C substitutions and HER2 activating alterations, which are likely to enter NSCLC guidelines upon the approval of the corresponding drugs. In addition to genetic examination, NSCLCs are usually subjected to the analysis of PD-L1 protein expression in order to direct the use of immune checkpoint inhibitors. Comprehensive NSCLC testing for multiple predictive markers requires the analysis of distinct biological molecules (DNA, RNA, proteins) and, therefore, the involvement of different analytical platforms (PCR, DNA sequencing, immunohistochemistry, FISH). There are ongoing efforts aimed at the integration of multiple NSCLC molecular assays into a single diagnostic pipeline.
Collapse
Affiliation(s)
- Evgeny N Imyanitov
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, St.-Petersburg, 197758, Russia; Department of Medical Genetics, St.-Petersburg Pediatric Medical University, St.-Petersburg, 194100, Russia; Department of Oncology, I.I. Mechnikov North-Western Medical University, St.-Petersburg, 195067, Russia.
| | - Aglaya G Iyevleva
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, St.-Petersburg, 197758, Russia; Department of Medical Genetics, St.-Petersburg Pediatric Medical University, St.-Petersburg, 194100, Russia
| | - Evgeny V Levchenko
- Department of Oncology, I.I. Mechnikov North-Western Medical University, St.-Petersburg, 195067, Russia; Department of Thoracic Oncology, N.N. Petrov Institute of Oncology, St.-Petersburg, 197758, Russia
| |
Collapse
|
20
|
Baglivo S, Ludovini V, Moretti R, Bellezza G, Sidoni A, Roila F, Metro G. RET Rearrangement as a Predictor of Unresponsiveness to Immunotherapy in Non-Small Cell Lung Cancer: Report of Two Cases with Review of the Literature. Oncol Ther 2020; 8:333-339. [PMID: 32700042 PMCID: PMC7683679 DOI: 10.1007/s40487-020-00116-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Indexed: 11/29/2022] Open
Abstract
Patients with epidermal growth factor receptor and anaplastic lymphoma kinase positive non-small cell lung cancer (NSCLC) generally respond poorly to treatment with immune checkpoint inhibitors such as anti-programmed cell death-1 (PD-1) or anti-programmed cell death ligand-1 (PD-L1) given with or without anti-cytotoxic T lymphocyte antigen-4 (CTLA-4) drugs. However, the efficacy of immunotherapy in patients with oncogene-addicted NSCLC harboring minor drivers, such as fusions in the rearranged during transfection (RET) gene, is still unclear. Here we describe two patients with RET-positive advanced NSCLC with PD-L1 expression ≥ 50% who developed progressive disease during first-line treatment with the anti-PD-1 agent pembrolizumab. In particular, while patient 2 was immediately switched to treatment with a selective RET inhibitor within the setting of a clinical trial, patient 1 responded to cytotoxic chemotherapy delivered at the time of progression while on pembrolizumab. These cases of NSCLC are discussed in the context of current literature, which seems to support our observation that patients with RET-positive NSCLC are unlikely to benefit from immunotherapy. Therefore, we suggest that for RET-positive patients with PD-L1 ≥ 50%, consideration should be given to upfront treatment approaches other than single-agent immunotherapy, namely selective RET inhibitors (if available) or regimens including cytotoxic chemotherapy.
Collapse
Affiliation(s)
- Sara Baglivo
- Laboratory of Oncology, Medical Oncology, Santa Maria della Misericordia Hospital, Azienda Ospedaliera di Perugia, Perugia, Italy
| | - Vienna Ludovini
- Laboratory of Oncology, Medical Oncology, Santa Maria della Misericordia Hospital, Azienda Ospedaliera di Perugia, Perugia, Italy
| | - Riccardo Moretti
- Department of Radiology, Santa Maria della Misericordia Hospital, Azienda Ospedaliera di Perugia, Perugia, Italy
| | - Guido Bellezza
- Department of Experimental Medicine, Division of Pathology and Histology, University of Perugia Medical School, Perugia, Italy
| | - Angelo Sidoni
- Department of Experimental Medicine, Division of Pathology and Histology, University of Perugia Medical School, Perugia, Italy
| | - Fausto Roila
- Medical Oncology, Santa Maria della Misericordia Hospital, Azienda Ospedaliera di Perugia, Perugia, Italy
| | - Giulio Metro
- Medical Oncology, Santa Maria della Misericordia Hospital, Azienda Ospedaliera di Perugia, Perugia, Italy.
| |
Collapse
|
21
|
Pelizzari G, Cortiula F, Giavarra M, Bartoletti M, Lisanti C, Buoro V, Cattaneo M, Rossetto C, Rizzato S, Puglisi F, Macerelli M, Fasola G, Follador A. Platinum-Based Chemotherapy in Older Patients with Non-Small Cell Lung Cancer: What to Expect in the Real World. Drugs Aging 2020; 37:677-689. [PMID: 32681401 DOI: 10.1007/s40266-020-00785-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND The role of platinum-based chemotherapy (PBC) for the treatment of older patients with non-small cell lung cancer (NSCLC) is still a matter of debate, despite the advent of immunotherapy. OBJECTIVE The aim of the study was to identify factors associated with first-line PBC prescription and, secondly, to evaluate the impact of first-line PBC on survival, treatment intensity, risk of hospitalization, and subsequent treatments. PATIENTS AND METHODS We reviewed a consecutive series of 474 older patients (age ≥ 70 years) diagnosed with stage IIIB-IV NSCLC at the Department of Oncology, University Hospital of Udine, Italy from January 2009 to March 2017. RESULTS Overall, 198 patients were deemed eligible, and 65.2% received a PBC. At multivariate analysis, older age was the only factor associated with PBC prescription. In the whole cohort, 46 patients (23.2%) were hospitalized for chemotherapy-related toxicity. Both PBC prescription (odds ratio [OR] 2.23, 95% confidence interval [CI] 1.02-4.87, p = 0.04) and tumor burden (OR 2.39, 95% CI 1.07-5.32, p = 0.03) emerged as independent risk factors for hospitalization. Moving to significant predictors of patterns of care, Eastern Cooperative Oncology Group (ECOG) performance status > 0 was associated with greater risk of first-line failure (OR 2.20, 95% CI 1.15-4.20, p = 0.02), while bone metastases (OR 0.29, 95% CI 0.12-0.69, p = 0.005) and a Charlson Comorbidity Index score ≥ 3 (OR 0.40, 95% CI 0.19-0.84, p = 0.016) independently predicted lower probability of receiving second-line therapy. Remarkably, PBC did not significantly impact overall survival (hazard ratio [HR] 0.83, 95% CI 0.61-1.14, p = 0.24) and progression-free survival (HR 0.95, 95% CI 0.70-1.28, p = 0.73) compared to single-agent chemotherapy (SAC). However, according to an exploratory landmark analysis, patients who received four cycles of treatment or maintenance therapy experienced prolonged overall survival, regardless of PBC use. CONCLUSIONS This study evaluated the real-world use of PBC in older patients with NSCLC, offering an insight into the determinants of its prescription and the pattern of care of these patients. Of note, PBC use was associated with a higher likelihood of hospitalization for chemotherapy-related toxicity, with no benefit on survival compared to SAC.
Collapse
Affiliation(s)
- Giacomo Pelizzari
- Department of Medicine (DAME), University of Udine, 33100, Udine, Italy. .,Department of Oncology, Santa Maria Della Misericordia Hospital (ASUFC), 33100, Udine, Italy.
| | - Francesco Cortiula
- Department of Medicine (DAME), University of Udine, 33100, Udine, Italy.,Department of Oncology, Santa Maria Della Misericordia Hospital (ASUFC), 33100, Udine, Italy
| | - Marco Giavarra
- Department of Medicine (DAME), University of Udine, 33100, Udine, Italy.,Department of Oncology, Santa Maria Della Misericordia Hospital (ASUFC), 33100, Udine, Italy
| | - Michele Bartoletti
- Department of Medicine (DAME), University of Udine, 33100, Udine, Italy.,Department of Oncology, Santa Maria Della Misericordia Hospital (ASUFC), 33100, Udine, Italy
| | - Camilla Lisanti
- Department of Medicine (DAME), University of Udine, 33100, Udine, Italy.,Department of Oncology, Santa Maria Della Misericordia Hospital (ASUFC), 33100, Udine, Italy
| | - Vanessa Buoro
- Department of Medicine (DAME), University of Udine, 33100, Udine, Italy.,Department of Oncology, Santa Maria Della Misericordia Hospital (ASUFC), 33100, Udine, Italy
| | - Monica Cattaneo
- Department of Medicine (DAME), University of Udine, 33100, Udine, Italy.,Department of Oncology, Santa Maria Della Misericordia Hospital (ASUFC), 33100, Udine, Italy
| | - Ciro Rossetto
- Department of Oncology, Santa Maria Della Misericordia Hospital (ASUFC), 33100, Udine, Italy
| | - Simona Rizzato
- Department of Oncology, Santa Maria Della Misericordia Hospital (ASUFC), 33100, Udine, Italy
| | - Fabio Puglisi
- Department of Medicine (DAME), University of Udine, 33100, Udine, Italy.,Dipartimento Di Oncologia Medica, Oncologia Medica E Prevenzione Oncologica, Centro Di Riferimento Oncologico Di Aviano (CRO), IRCCS, 33081, Aviano, Italy
| | - Marianna Macerelli
- Department of Oncology, Santa Maria Della Misericordia Hospital (ASUFC), 33100, Udine, Italy
| | - Gianpiero Fasola
- Department of Oncology, Santa Maria Della Misericordia Hospital (ASUFC), 33100, Udine, Italy
| | - Alessandro Follador
- Department of Oncology, Santa Maria Della Misericordia Hospital (ASUFC), 33100, Udine, Italy
| |
Collapse
|
22
|
Perioperative considerations for neoadjuvant immunotherapy in non–small cell lung cancer. J Thorac Cardiovasc Surg 2020; 160:1376-1382. [DOI: 10.1016/j.jtcvs.2020.05.119] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 05/12/2020] [Accepted: 05/13/2020] [Indexed: 11/21/2022]
|
23
|
Swan K, Dougherty KC, Myers SW. Somatic Testing and Germline Genetic Status: Implications for Cancer Treatment Decisions and Genetic Counseling. CURRENT GENETIC MEDICINE REPORTS 2020. [DOI: 10.1007/s40142-020-00192-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
24
|
Peng S, Ying AF, Tai BC, Soo RA. A meta-analysis on immune checkpoint inhibitor efficacy for advanced non-small cell lung cancer between East Asians versus non-East Asians. Transl Lung Cancer Res 2020; 9:1124-1137. [PMID: 32953491 PMCID: PMC7481594 DOI: 10.21037/tlcr-20-246] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 06/11/2020] [Indexed: 12/26/2022]
Abstract
BACKGROUND We conducted a meta-analysis to assess the efficacy of immune checkpoint inhibitors (ICIs) (PD-1/L1 and CTLA-4 inhibitors) in first and subsequent lines in East Asians and non-East Asians. METHODS We searched PubMed-MEDLINE, Embase and Scopus, from inception to 20 Sep 2019, and reviewed major conferences' abstracts, for randomised controlled trials of ICI in advanced-stage NSCLC (Stage IIIB or IV) without EGFR mutation that reported hazard ratios (HRs) stratified by geographical region including the region "Asia" or "East Asia". The primary outcome measures were overall survival (OS) and progression-free survival (PFS). The pooled HR and its 95% confidence interval (CI) for OS and PFS in East Asians and non-East Asians were calculated using a random effect model and the difference compared using an interaction test. RESULTS A total of 5,465 patients from 7 randomised controlled trials involving CTLA-4 and/or PD-1/L1 inhibitors were included, with 1,740 (32%) East Asians and 3,725 (68%) non-East Asians. ICI was associated with an improvement in OS and PFS for both East Asian (OS HR, 0.74; 95% CI, 0.65-0.85; PFS HR, 0.56; 95% CI, 0.40-0.79) and non-East Asian patients (OS HR, 0.78; 95% CI, 0.72-0.85; PFS HR, 0.69; 95% CI, 0.56-0.85), with no significant difference between the two groups (Pinteraction=0.55 for OS; Pinteraction=0.33 for PFS). Subgroup analyses showed a statistically significant superior PFS (but not OS) for East Asians than non-East Asians in trials that used immune checkpoint inhibitor in the first-line treatment (Pinteraction=0.02). No significant regional difference was found in further subgroups of pure ICI and combination of ICI with chemotherapy. CONCLUSIONS There is no significant difference in response to ICI between East Asians and non-East Asians with advanced stage NSCLC, and the statistically significant subgroup difference in PFS in the first line use of ICI may not be clinically significant.
Collapse
Affiliation(s)
- Siyu Peng
- Department of Medicine, National University Health System, Singapore, Singapore
| | - Ariel Fangting Ying
- Health Services and System Research, Duke-NUS Medical School, Singapore, Singapore
| | - Bee Choo Tai
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Ross Andrew Soo
- Department of Haematology-Oncology, National University Cancer Institute Singapore, National University Health System, Singapore, Singapore
- Cancer Science Institute, Singapore, Singapore
| |
Collapse
|
25
|
Xu LB, Zhao ZG, Xu SF, Zhang XX, Liu T, Jing CY, Zhang SG, Yu SJ. The landscape of gene mutations and clinical significance of tumor mutation burden in patients with soft tissue sarcoma who underwent surgical resection and received conventional adjuvant therapy. Int J Biol Markers 2020; 35:14-22. [PMID: 32520634 DOI: 10.1177/1724600820925095] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND The aim of this study was to evaluate the landscape of gene mutations and the clinical significance of tumor mutation burden (TMB) in patients with soft tissue sarcoma who underwent surgical resection and received conventional adjuvant therapy. METHODS A total of 68 patients with soft tissue sarcoma were included. Postoperative tumor tissue specimens from the patients were collected for DNA extraction. Targeted next-generation sequencing of cancer-relevant genes was performed for the detection of gene mutations and the analysis of TMB. Univariate analysis between TMB status and prognosis was carried out using the Kaplan-Meier survival analysis, and multivariate analysis was adjusted by the Cox regression model. RESULTS No specific genetic mutations associated with soft tissue sarcoma were found. The mutation frequency of TP53, PIK3C2G, NCOR1, and KRAS of the 68 patients with soft tissue sarcoma were observed in 19 cases (27.94%), 15 cases (22.06%), 14 cases (20.59%), and 14 cases (20.59%), respectively. With regard to the analysis of TMB, the overall TMB of the 68 patients with soft tissue sarcoma was relatively low (median: 2.05 per Mb (range: 0∼15.5 per Mb)). Subsequently, TMB status was divided into TMB-Low and TMB-Middle according to the median TMB. Patients with TMB-Low and TMB-Middle were 37 cases (54.41%) and 31 cases (45.59%), respectively. Overall survival analysis indicated that the median overall survival of patients with TMB-Low and TMB-Middle was not reached, and 4.5 years, respectively (P=0.015). CONCLUSION This study characterizes the genetic background of patients with STS soft tissue sarcoma. The TMB was of clinical significance for patients with soft tissue sarcoma who underwent surgical resection and received conventional adjuvant therapy.
Collapse
Affiliation(s)
- Li-Bin Xu
- Department of Orthopedic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhen-Guo Zhao
- Department of Orthopedic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Song-Feng Xu
- Department of Orthopedic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xin-Xin Zhang
- Department of Orthopedic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ting Liu
- Department of Orthopedic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chang-You Jing
- Department of Orthopedic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shu-Guang Zhang
- Department of Orthopedic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Sheng-Ji Yu
- Department of Orthopedic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
26
|
Kauffmann-Guerrero D, Tufman A, Kahnert K, Bollmann BA, Reu S, Syunyaeva Z, Schneider C, Manapov F, Huber RM, Golpon H. Response to Checkpoint Inhibition in Non-Small Cell Lung Cancer with Molecular Driver Alterations. Oncol Res Treat 2020; 43:289-298. [PMID: 32268332 DOI: 10.1159/000506842] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 02/26/2020] [Indexed: 11/19/2022]
Abstract
AIMS Non-small cell lung cancer (NSCLC) patients with EGFR mutations do not respond well to checkpoint inhibitors. However, little is known about the activity of immunotherapy in NSCLC with other driver mutations. The increasing use of next-generation sequencing (NGS) leads to molecular findings that face the clinician with problems while choosing the best treatment. This study aims at analyzing response of NSCLC with driver mutations to immunotherapy. PATIENTS AND METHODS We retrospectively included 84 NSCLC patients diagnosed and treated at 2 German tertiary-care lung cancer centers using NGS and treatment with immunotherapy. Response to immunotherapy was analyzed in correlation to molecular findings. RESULTS 51 patients harbored at least 1 driver mutation. PIK3CA, EGFR, and STK11 mutations did not respond to immunotherapy. KRAS, TP53, and MET exon 14 skipping mutations responded well. One patient with NF-1 mutation showed durable response. CONCLUSIONS Molecular testing may be of use in guiding treatment decision making in NSCLC.
Collapse
Affiliation(s)
- Diego Kauffmann-Guerrero
- Division of Respiratory Medicine and Thoracic Oncology, Department of Internal Medicine V, Thoracic Oncology Center Munich, University of Munich (LMU), Munich, Germany,
| | - Amanda Tufman
- Division of Respiratory Medicine and Thoracic Oncology, Department of Internal Medicine V, Thoracic Oncology Center Munich, University of Munich (LMU), Munich, Germany
| | - Kathrin Kahnert
- Division of Respiratory Medicine and Thoracic Oncology, Department of Internal Medicine V, Thoracic Oncology Center Munich, University of Munich (LMU), Munich, Germany
| | | | - Simone Reu
- Institute for Pathology, University of Munich, Munich, Germany
| | - Zulfiya Syunyaeva
- Division of Respiratory Medicine and Thoracic Oncology, Department of Internal Medicine V, Thoracic Oncology Center Munich, University of Munich (LMU), Munich, Germany
| | | | - Farkhad Manapov
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Rudolf M Huber
- Division of Respiratory Medicine and Thoracic Oncology, Department of Internal Medicine V, Thoracic Oncology Center Munich, University of Munich (LMU), Munich, Germany
| | - Heiko Golpon
- Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany
| |
Collapse
|
27
|
Filipovic A, Miller G, Bolen J. Progress Toward Identifying Exact Proxies for Predicting Response to Immunotherapies. Front Cell Dev Biol 2020; 8:155. [PMID: 32258034 PMCID: PMC7092703 DOI: 10.3389/fcell.2020.00155] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 02/26/2020] [Indexed: 12/14/2022] Open
Abstract
Clinical value and utility of checkpoint inhibitors, a drug class targeting adaptive immune suppression pathways (PD-1, PDL-1, and CTLA-4), is growing rapidly and maintains status of a landmark achievement in oncology. Their efficacy has transformed life expectancy in multiple deadly cancer types (melanoma, lung cancer, renal/urothelial carcinoma, certain colorectal cancers, lymphomas, etc.). Despite significant clinical development efforts, therapeutic indication of approved checkpoint inhibitors are not as wide as the oncology community and patients would like them to be, potentially bringing into question their universal efficacy across tumor histologies. With the main goal of expanding immunotherapy applications, identifying of biomarkers to accurately predict therapeutic response and treatment related side-effects are a paramount need in the field. Specificities surrounding checkpoint inhibitors in clinic, such as unexpected tumor response patterns (pseudo- and hyper-progression), late responders, as well as specific immune mediated toxicities, complicate the management of patients. They stem from the complexities and dynamics of the tumor/host immune interactions, as well as baseline tumor biology. Search for clinically effective biomarkers therefore calls for a holistic approach, rather than implementation of a single analyte. The goal is to achieve dynamic and comprehensive acquisition, analyses and interpretation of immunological and biologic information about the tumor and the immune system, and to compute these parameters into an actionable, maximally predictive value at the individual patient level. Limitation delaying swift incorporation of validated immuno-oncology biomarkers span from standardized biospecimens acquisition and processing, selection of proficient biomarker discovery and validation methods, to establishing multidisciplinary consortiums and data sharing platforms. Multi-disciplinary efforts have already yielded some approved (PDL-1 and MSI-status) and other advanced tests (TMB, neoantigen pattern, and TIL infiltration rate). Importantly, clinical trial taskforces now recognize the imperative of the biomarker-driven trial design and execution, to enable translating biomarker discoveries into the clinical setting. This will ensure we utilize the “conspiracy” between the peripheral and intra-tumoral dynamic markers in shaping responses to checkpoint blockade, for the ultimate patient benefit.
Collapse
Affiliation(s)
| | - George Miller
- New York University School of Medicine, New York, NY, United States
| | | |
Collapse
|
28
|
Bashraheel SS, Domling A, Goda SK. Update on targeted cancer therapies, single or in combination, and their fine tuning for precision medicine. Biomed Pharmacother 2020; 125:110009. [PMID: 32106381 DOI: 10.1016/j.biopha.2020.110009] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Revised: 02/04/2020] [Accepted: 02/12/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Until recently, patients who have the same type and stage of cancer all receive the same treatment. It has been established, however, that individuals with the same disease respond differently to the same therapy. Further, each tumor undergoes genetic changes that cause cancer to grow and metastasize. The changes that occur in one person's cancer may not occur in others with the same cancer type. These differences also lead to different responses to treatment. Precision medicine, also known as personalized medicine, is a strategy that allows the selection of a treatment based on the patient's genetic makeup. In the case of cancer, the treatment is tailored to take into account the genetic changes that may occur in an individual's tumor. Precision medicine, therefore, could be defined in terms of the targets involved in targeted therapy. METHODS A literature search in electronic data bases using keywords "cancer targeted therapy, personalized medicine and cancer combination therapies" was conducted to include papers from 2010 to June 2019. RESULTS Recent developments in strategies of targeted cancer therapy were reported. Specifically, on the two types of targeted therapy; first, immune-based therapy such as the use of immune checkpoint inhibitors (ICIs), immune cytokines, tumor-targeted superantigens (TTS) and ligand targeted therapeutics (LTTs). The second strategy deals with enzyme/small molecules-based therapies, such as the use of a proteolysis targeting chimera (PROTAC), antibody-drug conjugates (ADC) and antibody-directed enzyme prodrug therapy (ADEPT). The precise targeting of the drug to the gene or protein under attack was also investigated, in other words, how precision medicine can be used to tailor treatments. CONCLUSION The conventional therapeutic paradigm for cancer and other diseases has focused on a single type of intervention for all patients. However, a large literature in oncology supports the therapeutic benefits of a precision medicine approach to therapy as well as combination therapies.
Collapse
Affiliation(s)
- Sara S Bashraheel
- Protein Engineering Unit, Life and Science Research Department, Anti-Doping Lab-Qatar (ADLQ), Doha, Qatar; Drug Design Group, Department of Pharmacy, University of Groningen, Groningen, Netherlands
| | - Alexander Domling
- Drug Design Group, Department of Pharmacy, University of Groningen, Groningen, Netherlands
| | - Sayed K Goda
- Cairo University, Faculty of Science, Chemistry Department, Giza, Egypt.
| |
Collapse
|
29
|
De Giglio A, Nuvola G, Dall'Olio FG. Carboplatin, nab-paclitaxel plus atezolizumab in IMpower 130 trial: new weapons beyond controversies. Transl Cancer Res 2019; 8:E18-E23. [PMID: 35117058 PMCID: PMC8798162 DOI: 10.21037/tcr.2019.12.69] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 12/19/2019] [Indexed: 11/06/2022]
Affiliation(s)
- Andrea De Giglio
- Department of Specialized, Experimental and Diagnostic Medicine, S.Orsola-Malpighi University Hospital, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Giacomo Nuvola
- Department of Specialized, Experimental and Diagnostic Medicine, S.Orsola-Malpighi University Hospital, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Filippo Gustavo Dall'Olio
- Department of Specialized, Experimental and Diagnostic Medicine, S.Orsola-Malpighi University Hospital, Alma Mater Studiorum University of Bologna, Bologna, Italy
| |
Collapse
|