Abstract
Original language | English |
---|---|
Journal | J. Immunother. Cancer |
Volume | 8 |
Issue number | 2 |
DOIs | |
Publication status | Published - 2020 |
Keywords
- immunotherapy
- acetylsalicylic acid
- antibiotic agent
- anticoagulant agent
- antidiabetic agent
- antilipemic agent
- atezolizumab
- beta adrenergic receptor blocking agent
- calcium channel blocking agent
- corticosteroid
- immunosuppressive agent
- metformin
- nivolumab
- nonsteroid antiinflammatory agent
- nystatin
- opiate
- pembrolizumab
- programmed death 1 ligand 1
- programmed death 1 receptor
- proton pump inhibitor
- serotonin 1 antagonist
- adult
- aged
- Article
- body mass
- cancer immunotherapy
- cancer staging
- clinical evaluation
- clinical outcome
- clinical practice
- disease burden
- disease exacerbation
- drug cost
- drug efficacy
- Eastern Cooperative Oncology Group Performance Status
- female
- gastritis
- gastroesophageal reflux
- human
- inflammation
- lung cancer
- major clinical study
- male
- melanoma
- multicenter study
- non small cell lung cancer
- obesity
- observational study
- overall survival
- palliative therapy
- prescription
- priority journal
- progression free survival
- prophylaxis
- retrospective study
- screening test
- stomach acid
- tumor associated leukocyte
- tumor growth
- tumor-related gene
Fingerprint
Dive into the research topics of 'Integrated analysis of concomitant medications and oncological outcomes from PD-1/PD-L1 checkpoint inhibitors in clinical practice: Journal for ImmunoTherapy of Cancer'. Together they form a unique fingerprint.Cite this
- APA
- Standard
- Harvard
- Vancouver
- Author
- BIBTEX
- RIS
Integrated analysis of concomitant medications and oncological outcomes from PD-1/PD-L1 checkpoint inhibitors in clinical practice : Journal for ImmunoTherapy of Cancer. / Cortellini, A.; Tucci, M.; Adamo, V. et al.
In: J. Immunother. Cancer, Vol. 8, No. 2, 2020.Research output: Contribution to journal › Article › peer-review
}
TY - JOUR
T1 - Integrated analysis of concomitant medications and oncological outcomes from PD-1/PD-L1 checkpoint inhibitors in clinical practice
T2 - Journal for ImmunoTherapy of Cancer
AU - Cortellini, A.
AU - Tucci, M.
AU - Adamo, V.
AU - Stucci, L.S.
AU - Russo, A.
AU - Tanda, E.T.
AU - Spagnolo, F.
AU - Rastelli, F.
AU - Bisonni, R.
AU - Santini, D.
AU - Russano, M.
AU - Anesi, C.
AU - Giusti, R.
AU - Filetti, M.
AU - Marchetti, P.
AU - Botticelli, A.
AU - Gelibter, A.
AU - Occhipinti, M.A.
AU - Marconcini, R.
AU - Vitale, M.G.
AU - Nicolardi, L.
AU - Chiari, R.
AU - Bareggi, C.
AU - Nigro, O.
AU - Tuzi, A.
AU - De Tursi, M.
AU - Petragnani, N.
AU - Pala, L.
AU - Bracarda, S.
AU - MacRini, S.
AU - Inno, A.
AU - Zoratto, F.
AU - Veltri, E.
AU - DI Cocco, B.
AU - Mallardo, D.
AU - Pinato, D.J.
AU - Porzio, G.
AU - Ficorella, C.
AU - Ascierto, P.A.
N1 - Cited By :1 Export Date: 2 March 2021 Correspondence Address: Cortellini, A.; Department of Biotechnology and Applied Clinical Sciences, Italy; email: alessiocortellini@gmail.com Chemicals/CAS: acetylsalicylic acid, 493-53-8, 50-78-2, 53663-74-4, 53664-49-6, 63781-77-1; atezolizumab, 1380723-44-3; metformin, 1115-70-4, 657-24-9; nivolumab, 946414-94-4; nystatin, 1400-61-9, 34786-70-4, 62997-67-5; opiate, 53663-61-9, 8002-76-4, 8008-60-4; pembrolizumab, 1374853-91-4 Funding details: Pfizer Funding details: Novartis Funding details: Wellcome Trust, WT, PS3416 Funding details: Imperial Experimental Cancer Medicine Centre, ECMC Funding details: British Microcirculation Society, BMS Funding details: Astellas Pharma Funding details: NIHR Imperial Biomedical Research Centre, BRC Funding details: Ipsen Funding details: Consorzio Nazionale Interuniversitario per le Telecomunicazioni, CNIT Funding text 1: Funding This work was supported by the Consorzio Interuniversitario Nazionale per la Bio-Oncologia (CINBO). DP is supported by grant funding from the Wellcome Trust Strategic Fund (PS3416) and from the NIHR Imperial Biomedical Research Center (BRC) ITMAT Push for Impact Scheme 2019 and acknowledges infrastructural support by the Cancer Research UK Imperial Center and the Imperial Experimental Cancer Medicine Center (ECMC). Funding text 2: Competing interests AC received speaker fees and grant consultancies from Roche, MSD, BMS, AstraZeneca, Novartis, Astellas. RG received speaker fees and grant consultancies from AstraZeneca and Roche. MGV received speaker fees, grant consultancies and travel support from BMS, Ipsen, Novartis, Pfizer, Astellas, Jansen and Pierre-Fabre. AR received grant consultancies from AstraZeneca and MSD. RM received grant consultancies from Pierre-Fabre, MSD, Incyte, BMS, and Roche. FS received speaker fees and grant consultancies from Roche, Novartis, BMS, MSD, Pierre-Fabre, Sanofi, Merck and Sunpharma. DP received lecture fees from ViiV Healthcare, Bayer Healthcare and travel expenses from BMS and Bayer Healthcare; consulting fees for Mina Therapeutics, EISAI, Roche, Astra Zeneca; received research funding (to institution) from MSD, BMS. PAA received speaker fees and grant consultancies from BMS, Roche-Genentech, MSD, Dohme, Array, Novartis, Merck-Serono, Pierre-Fabre, Incyte, New Link Genetics, Genmab, Medimmune, AstraZeneca, Syndax, SunPharma, Sanofi, Idera, Ultimovacs, Sandoz, Immunocore, 4SC, Alkermes, Italfarmaco, Nektar, Boehringer-Ingelheim; he also received research funds from BMS, Roche-Genentech, Array. References: Scripture, C.D., Figg, W.D., Drug interactions in cancer therapy (2006) Nat Rev Cancer, 6, pp. 546-558. , [published correction appears in Nat Rev Cancer. 2006 Sep;6(9):741]; Hussain, N., Naeem, M., Pinato, D.J., Concomitant medications and immune checkpoint inhibitor therapy for cancer: Causation or association? (2020) Hum Vaccin Immunother, pp. 1-7; Gopalakrishnan, V., Helmink, B.A., Spencer, C.N., The influence of the gut microbiome on cancer, immunity, and cancer immunotherapy (2018) Cancer Cell, 33, pp. 570-580; Malmberg, K.-J., Effective immunotherapy against cancer: A question of overcoming immune suppression and immune escape? (2004) Cancer Immunol Immunother, 53, pp. 879-892; Arbour, K.C., Mezquita, L., Long, N., Impact of baseline steroids on efficacy of programmed cell death-1 and programmed Death-Ligand 1 blockade in patients with non-small-cell lung cancer (2018) J Clin Oncol, 36, pp. 2872-2878; Ricciuti, B., Dahlberg, S.E., Adeni, A., Immune checkpoint inhibitor outcomes for patients with non-small-cell lung cancer receiving baseline corticosteroids for palliative versus Nonpalliative indications (2019) J Clin Oncol, 37, pp. 1927-1934; Petrelli, F., Signorelli, D., Ghidini, M., Association of steroids use with survival in patients treated with immune checkpoint inhibitors: A systematic review and meta-analysis (2020) Cancers, 12, p. 546; Derosa, L., Hellmann, M.D., Spaziano, M., Negative association of antibiotics on clinical activity of immune checkpoint inhibitors in patients with advanced renal cell and non-small-cell lung cancer (2018) Ann Oncol, 29, pp. 1437-1444; Chalabi, M., Cardona, A., Nagarkar, D.R., Efficacy of chemotherapy and atezolizumab in patients with non-small-cell lung cancer receiving antibiotics and proton pump inhibitors: Pooled post hoc analyses of the oak and poplar trials (2020) Ann Oncol, 31, pp. 525-531; Pinato, D.J., Howlett, S., Ottaviani, D., Association of prior antibiotic treatment with survival and response to immune checkpoint inhibitor therapy in patients with cancer (2019) JAMA Oncol, 5, pp. 1774-1778; Homicsko, K., Richtig, G., Tuchmann, F., Proton pump inhibitors negatively impact survival of PD-1 inhibitor based therapies in metastatic melanoma patients (2018) Ann Oncol, 29, p. x40; Wang, D.Y., McQuade, J.L., Rai, R.R., The impact of nonsteroidal anti-inflammatory drugs, beta blockers, and metformin on the efficacy of anti-PD-1 therapy in advanced melanoma (2020) Oncologist, 25, pp. e602-e605; Gandhi, S., Pandey, M., Ammannagari, N., Impact of concomitant medication use and immune-related adverse events on response to immune checkpoint inhibitors (2020) Immunotherapy, 12, pp. 141-149; Hakozaki, T., Hosomi, Y., Shimizu, A., Polypharmacy as a prognostic factor in older patients with advanced non-small-cell lung cancer treated with anti-PD-1/PD-L1 antibody-based immunotherapy (2020) J Cancer Res Clin Oncol, 146, pp. 2659-2668; Cortellini, A., Bersanelli, M., Buti, S., A multicenter study of body mass index in cancer patients treated with anti-PD-1/PD-L1 immune checkpoint inhibitors: When overweight becomes favorable (2019) J Immunother Cancer, 7, p. 57; Cortellini, A., Bersanelli, M., Santini, D., Another side of the association between body mass index (BMI) and clinical outcomes of cancer patients receiving programmed cell death protein-1 (PD-1)/ Programmed cell death-ligand 1 (PD-L1) checkpoint inhibitors: A multicentre analysis of immune-related adverse events (2020) Eur J Cancer, 128, pp. 17-26; Cortellini, A., Buti, S., Bersanelli, M., Evaluating the role of family history of cancer and diagnosis of multiple neoplasms in cancer patients receiving PD-1/PD-L1 checkpoint inhibitors: The multicenter FAMI-L1 study (2020) Oncoimmunology, 9, p. 1710389; Cortellini, A., Vitale, M.G., De Galitiis, F., Early fatigue in cancer patients receiving PD-1/PD-L1 checkpoint inhibitors: An insight from clinical practice (2019) J Transl Med, 17, p. 376; Cortellini, A., Buti, S., Santini, D., Clinical outcomes of patients with advanced cancer and pre-existing autoimmune diseases treated with Anti-Programmed death-1 immunotherapy: A real-world transverse study (2019) Oncologist, 24, pp. e327-e337; Cortellini, A., Chiari, R., Ricciuti, B., Correlations between the immune-related adverse events spectrum and efficacy of anti-PD1 immunotherapy in NSCLC patients (2019) Clin Lung Cancer, 20, pp. 237-247; Eisenhauer, E.A., Therasse, P., Bogaerts, J., New response evaluation criteria in solid tumours: Revised RECIST guideline (version 1 (2009) 1). Eur J Cancer, 45, pp. 228-247; Woolley, K.K., How variables uncorrelated with the dependent variable can actually make excellent predictors: The important suppressor variable case (1997) Southwest Educational Research Association Annual Meeting Proceedings, , https://eric.ed.gov/?id=ED407420[Accessed10Jun2020]; Thompson, F.T., Levine, D.U., Examples of easily Explainable suppressor variables in multiple regression research (1997) Multi Linear Regres Viewpoints, 24, pp. 11-13; "stopping Stepwise: Why Stepwise Selection is Bad and What You Should Use Instead", , https://towardsdatascience.com/stopping-stepwise-why-stepwiseselection-is-bad-and-what-you-should-use-instead-90818b3f52df, On towardsdatascience. Com, [Accessed 10 Jun 2020]; Minana, B., Cozar, J.M., Palou, J., Bladder cancer in Spain 2011: Population based study (2014) J Urol, 191, pp. 323-328; Ciocan, D., Barbe, C., Aubin, F., Distinctive features of melanoma and its management in elderly patients: A population-based study in France (2013) JAMA Dermatol, 149, pp. 1150-1157; Gridelli, C., Balducci, L., Ciardiello, F., Treatment of elderly patients with non-small-cell lung cancer: Results of an international expert panel meeting of the Italian association of thoracic oncology (2015) Clin Lung Cancer, 16, pp. 325-333; Azawi, N.H., Joergensen, S.M., Jensen, N.V., Trends in kidney cancer among the elderly in Denmark, 1980-2012 (2016) Acta Oncol, 55, pp. 79-84; Wu, T., Yang, L., Jiang, J., Chronic glucocorticoid treatment induced circadian clock disorder leads to lipid metabolism and gut microbiota alterations in rats (2018) Life Sci, 192, pp. 173-182; He, Z., Kong, X., Shao, T., Alterations of the gut microbiota associated with promoting efficacy of prednisone by Bromofuranone in MRL/lpr mice (2019) Front Microbiol, 10, p. 978; Li, Z.-Y., Fan, M.-B., Zhang, S.-L., Intestinal Metrnl released into the gut lumen acts as a local regulator for gut antimicrobial peptides (2016) Acta Pharmacol Sin, 37, pp. 1458-1466; Hagan, T., Cortese, M., Rouphael, N., Antibiotics-Driven gut microbiome perturbation alters immunity to vaccines in humans (2019) Cell, 178, pp. 1313e13-28e13; Bruno, G., Zaccari, P., Rocco, G., Proton pump inhibitors and dysbiosis: Current knowledge and aspects to be clarified (2019) World J Gastroenterol, 25, pp. 2706-2719; Jackson, M.A., Goodrich, J.K., Maxan, M.-E., Proton pump inhibitors alter the composition of the gut microbiota (2016) Gut, 65, pp. 749-756; Gao, C., Major, A., Rendon, D., Histamine H2 receptor-mediated suppression of intestinal inflammation by probiotic Lactobacillus reuteri (2015) MBio, 6, pp. e01358-e01415; Diebel, L.N., Liberati, D.M., Hall-Zimmerman, L., H2 blockers decrease gut mucus production and lead to barrier dysfunction in vitro (2011) Surgery, 150, pp. 736-743; Questions and Answers: NDMA Impurities in Ranitidine (Commonly Known As Zantac), , https://www.fda.gov/drugs/drug-safety-and-availability/questionsand-answers-ndma-impurities-ranitidine-commonly-known-zantac, U.S. Food and Drug Administration, [Accessed 14 Jun 2020]; EMA to Review Ranitidine Medicines following Detection of NDMA, , https://www.ema.europa.eu/en/news/ema-review-ranitidine-medicines-following-detection-ndma, European Medicines Agencies, [Accessed 14 Jun 2020]; Strobel, L., Johswich, K.O., Anticoagulants impact on innate immune responses and bacterial survival in whole blood models of Neisseria meningitidis infection (2018) Sci Rep, 8, p. 10225; Taylor, A., Revealing a brain-gut microbiome connection following chronic opioid treatment (2018) J Pain, 19, p. S1; Acharya, C., Betrapally, N.S., Gillevet, P.M., Chronic opioid use is associated with altered gut microbiota and predicts readmissions in patients with cirrhosis (2017) Aliment Pharmacol Ther, 45, pp. 319-331; Banerjee, S., Sindberg, G., Wang, F., Opioid-induced gut microbial disruption and bile dysregulation leads to gut barrier compromise and sustained systemic inflammation (2016) Mucosal Immunol, 9, pp. 1418-1428; Ren, M., Lotfipour, S., The role of the gut microbiome in opioid use (2020) Behav Pharmacol, 31, pp. 113-121; Dai, X., Yan, J., Fu, X., Aspirin inhibits cancer metastasis and angiogenesis via targeting heparanase (2017) Clin Cancer Res, 23, pp. 6267-6278; Algra, A.M., Rothwell, P.M., Effects of regular aspirin on long-term cancer incidence and metastasis: A systematic comparison of evidence from observational studies versus randomised trials (2012) Lancet Oncol, 13, pp. 518-527; Botti, G., Fratangelo, F., Cerrone, M., COX-2 expression positively correlates with PD-L1 expression in human melanoma cells (2017) J Transl Med, 15, p. 46; Zelenay, S., Van Der Veen, A.G., Bottcher, J.P., Cyclooxygenase-Dependent tumor growth through evasion of immunity (2015) Cell, 162, pp. 1257-1270; Ma, X., Bi, E., Lu, Y., Cholesterol Induces CD8+ T Cell Exhaustion in the Tumor Microenvironment (2019) Cell Metab, 30, pp. 143e5-56e5; Yang, W., Bai, Y., Xiong, Y., Potentiating the antitumour response of CD8(+) T cells by modulating cholesterol metabolism (2016) Nature, 531, pp. 651-655; Mok, E.H.K., Lee, T.K.W., The pivotal role of the dysregulation of cholesterol homeostasis in cancer: Implications for therapeutic targets (2020) Cancers, 12, p. E1410; Perrone, F., Minari, R., Bersanelli, M., The prognostic role of high blood cholesterol in advanced cancer patients treated with immune checkpoint inhibitors (2020) J Immunother, 43, pp. 196-203; De Giorgi, V., Grazzini, M., Benemei, S., Propranolol for off-label treatment of patients with melanoma: Results from a cohort study (2018) JAMA Oncol, 4, p. e172908; Kokolus, K.M., Zhang, Y., Sivik, J.M., Beta blocker use correlates with better overall survival in metastatic melanoma patients and improves the efficacy of immunotherapies in mice (2018) Oncoimmunology, 7, p. e1405205; Thiele, M., Wiest, R., Gluud, L.L., Can non-selective beta-blockers prevent hepatocellular carcinoma in patients with cirrhosis? (2013) Med Hypotheses, 81, pp. 871-874; Kichenadasse, G., Miners, J.O., Mangoni, A.A., Association between body mass index and overall survival with immune checkpoint inhibitor therapy for advanced non-small cell lung cancer (2020) JAMA Oncol, 6, p. e195241; Afzal, M.Z., Mercado, R.R., Shirai, K., Efficacy of metformin in combination with immune checkpoint inhibitors (anti-PD-1/anti-CTLA-4) in metastatic malignant melanoma (2018) J Immunother Cancer, 6, p. 64; Coutinho, A.E., Chapman, K.E., The anti-inflammatory and immunosuppressive effects of glucocorticoids, recent developments and mechanistic insights (2011) Mol Cell Endocrinol, 335, pp. 2-13; Luo, Y., Zheng, S.G., Hall of fame among pro-inflammatory cytokines: Interleukin-6 gene and its transcriptional regulation mechanisms (2016) Front Immunol, 7, p. 604; Giles, A.J., Hutchinson, M.-K.N.D., Sonnemann, H.M., Dexamethasone-induced immunosuppression: Mechanisms and implications for immunotherapy (2018) J Immunother Cancer, 6, p. 51; Biswas, S., Benedict, S.H., Lynch, S.G., Potential immunological consequences of pharmacological suppression of gastric acid production in patients with multiple sclerosis (2012) BMC Med, 10, p. 57; Hussain, M., Javeed, A., Ashraf, M., Aspirin and immune system (2012) Int Immunopharmacol, 12, pp. 10-20
PY - 2020
Y1 - 2020
N2 - Background Concomitant medications, such as steroids, proton pump inhibitors (PPI) and antibiotics, might affect clinical outcomes with immune checkpoint inhibitors. Methods We conducted a multicenter observational retrospective study aimed at evaluating the impact of concomitant medications on clinical outcomes, by weighing their associations with baseline clinical characteristics (including performance status, burden of disease and body mass index) and the underlying causes for their prescription. This analysis included consecutive stage IV patients with cancer, who underwent treatment with single agent antiprogrammed death-1/programmed death ligand-1 (PD-1/PD-L1) with standard doses and schedules at the medical oncology departments of 20 Italian institutions. Each medication taken at the immunotherapy initiation was screened and collected into key categories as follows: corticosteroids, antibiotics, gastric acid suppressants (including proton pump inhibitors - PPIs), statins and other lipid-lowering agents, aspirin, anticoagulants, non-steroidal anti-inflammatory drugs (NSAIDs), ACE inhibitors/Angiotensin II receptor blockers, calcium antagonists, β-blockers, metformin and other oral antidiabetics, opioids. Results From June 2014 to March 2020, 1012 patients were included in the analysis. Primary tumors were: non-small cell lung cancer (52.2%), melanoma (26%), renal cell carcinoma (18.3%) and others (3.6%). Baseline statins (HR 1.60 (95% CI 1.14 to 2.25), p=0.0064), aspirin (HR 1.47 (95% CI 1.04 to 2.08, p=0.0267) and β-blockers (HR 1.76 (95% CI 1.16 to 2.69), p=0.0080) were confirmed to be independently related to an increased objective response rate. Patients receiving cancer-related steroids (HR 1.72 (95% CI 1.43 to 2.07), p
AB - Background Concomitant medications, such as steroids, proton pump inhibitors (PPI) and antibiotics, might affect clinical outcomes with immune checkpoint inhibitors. Methods We conducted a multicenter observational retrospective study aimed at evaluating the impact of concomitant medications on clinical outcomes, by weighing their associations with baseline clinical characteristics (including performance status, burden of disease and body mass index) and the underlying causes for their prescription. This analysis included consecutive stage IV patients with cancer, who underwent treatment with single agent antiprogrammed death-1/programmed death ligand-1 (PD-1/PD-L1) with standard doses and schedules at the medical oncology departments of 20 Italian institutions. Each medication taken at the immunotherapy initiation was screened and collected into key categories as follows: corticosteroids, antibiotics, gastric acid suppressants (including proton pump inhibitors - PPIs), statins and other lipid-lowering agents, aspirin, anticoagulants, non-steroidal anti-inflammatory drugs (NSAIDs), ACE inhibitors/Angiotensin II receptor blockers, calcium antagonists, β-blockers, metformin and other oral antidiabetics, opioids. Results From June 2014 to March 2020, 1012 patients were included in the analysis. Primary tumors were: non-small cell lung cancer (52.2%), melanoma (26%), renal cell carcinoma (18.3%) and others (3.6%). Baseline statins (HR 1.60 (95% CI 1.14 to 2.25), p=0.0064), aspirin (HR 1.47 (95% CI 1.04 to 2.08, p=0.0267) and β-blockers (HR 1.76 (95% CI 1.16 to 2.69), p=0.0080) were confirmed to be independently related to an increased objective response rate. Patients receiving cancer-related steroids (HR 1.72 (95% CI 1.43 to 2.07), p
KW - immunotherapy
KW - acetylsalicylic acid
KW - antibiotic agent
KW - anticoagulant agent
KW - antidiabetic agent
KW - antilipemic agent
KW - atezolizumab
KW - beta adrenergic receptor blocking agent
KW - calcium channel blocking agent
KW - corticosteroid
KW - immunosuppressive agent
KW - metformin
KW - nivolumab
KW - nonsteroid antiinflammatory agent
KW - nystatin
KW - opiate
KW - pembrolizumab
KW - programmed death 1 ligand 1
KW - programmed death 1 receptor
KW - proton pump inhibitor
KW - serotonin 1 antagonist
KW - adult
KW - aged
KW - Article
KW - body mass
KW - cancer immunotherapy
KW - cancer staging
KW - clinical evaluation
KW - clinical outcome
KW - clinical practice
KW - disease burden
KW - disease exacerbation
KW - drug cost
KW - drug efficacy
KW - Eastern Cooperative Oncology Group Performance Status
KW - female
KW - gastritis
KW - gastroesophageal reflux
KW - human
KW - inflammation
KW - lung cancer
KW - major clinical study
KW - male
KW - melanoma
KW - multicenter study
KW - non small cell lung cancer
KW - obesity
KW - observational study
KW - overall survival
KW - palliative therapy
KW - prescription
KW - priority journal
KW - progression free survival
KW - prophylaxis
KW - retrospective study
KW - screening test
KW - stomach acid
KW - tumor associated leukocyte
KW - tumor growth
KW - tumor-related gene
U2 - 10.1136/jitc-2020-001361
DO - 10.1136/jitc-2020-001361
M3 - Article
SN - 2051-1426
VL - 8
JO - J. Immunother. Cancer
JF - J. Immunother. Cancer
IS - 2
ER -