Metabolic syndrome is independently associated with improved overall survival to first-line therapy with immune checkpoint inhibitors in non-small cell lung cancer

Background Many co-existing medical conditions may affect the outcome in patients treated with immune checkpoint inhibitors for advanced cancer. There is currently not any information on whether metabolic syndrome (MetS) impacts the clinical outcome in patients treated with immune checkpoint inhibitors (ICIs) for advanced non-small cell line cancer (NSCLC). Methods We carried out a single-center retrospective cohort study to determine the effects of MetS on first-line ICI therapy in patients with NSCLC. Results One hundred and eighteen consecutive adult patients who received first-line therapy with ICIs and had adequate medical record information for the determination of MetS status and clinical outcomes were included in the study. Twenty-one patients had MetS and 97 did not. There was no significant difference between the two groups in age, gender, smoking history, ECOG performance status, tumor histologic types, pre-therapy use of broad-spectrum antimicrobials, PD-L1 expression, pre-treatment neutrophil:lymphocyte ratio, or proportions of patients who received ICI monotherapy or chemoimmunotherapy. With a median follow-up of 9 months (range 0.5-67), MetS patients enjoyed significantly longer overall survival (HR 0.54, 95% CI: 0.31-0.92) (p = 0.02) but not progression-free survival. The improved outcome was only observed in patients who received ICI monotherapy and not chemoimmunotherapy. MetS predicted for higher probability of survival at 6 months (p = 0.043) and 12 months (p = 0.008). Multivariate analysis indicated that, in addition to the known adverse effects of use of broad-spectrum antimicrobials and the beneficial effects of PD-L1 (Programmed cell death-ligand 1) expression, MetS was independently associated with improved overall survival but not progression-free survival. Conclusions Our results suggest that MetS is an independent predictor of treatment outcome in patients who received first-line ICI monotherapy for NSCLC.

[1]  A. Madabhushi,et al.  Role of tumor infiltrating lymphocytes and spatial immune heterogeneity in sensitivity to PD-1 axis blockers in non-small cell lung cancer , 2022, Journal for ImmunoTherapy of Cancer.

[2]  K. Takayama,et al.  Impact of losing adipose tissue on outcomes from PD‐1/PD‐L1 inhibitor monotherapy in non‐small cell lung cancer , 2022, Thoracic cancer.

[3]  A. Addeo,et al.  Prognostic effect of body mass index in patients with advanced NSCLC treated with chemoimmunotherapy combinations , 2022, Journal for ImmunoTherapy of Cancer.

[4]  N. Segata,et al.  Intestinal Akkermansia muciniphila predicts clinical response to PD-1 blockade in patients with advanced non-small-cell lung cancer , 2022, Nature Medicine.

[5]  Qing Wang,et al.  Blood tumor mutation burden can predict the clinical response to immune checkpoint inhibitors in advanced non-small cell lung cancer patients , 2021, Cancer Immunology, Immunotherapy.

[6]  Tatsuo Kawai,et al.  Adipose Tissue Inflammation and Metabolic Dysfunction in Obesity. , 2020, American journal of physiology. Cell physiology.

[7]  Y. Hu,et al.  Pretreatment Neutrophil-to-Lymphocyte Ratio (NLR) May Predict the Outcomes of Advanced Non-small-cell Lung Cancer (NSCLC) Patients Treated With Immune Checkpoint Inhibitors (ICIs) , 2020, Frontiers in Oncology.

[8]  A. Rowland,et al.  Association Between Body Mass Index and Overall Survival With Immune Checkpoint Inhibitor Therapy for Advanced Non-Small Cell Lung Cancer. , 2019, JAMA oncology.

[9]  S. Krähenbühl,et al.  Predictive impact of antibiotics in patients with advanced non small-cell lung cancer receiving immune checkpoint inhibitors , 2019, Cancer Chemotherapy and Pharmacology.

[10]  S. Klein,et al.  Metabolically healthy obesity: facts and fantasies. , 2019, The Journal of clinical investigation.

[11]  H. Yao,et al.  Association of Survival and Immune-Related Biomarkers With Immunotherapy in Patients With Non–Small Cell Lung Cancer , 2019, JAMA network open.

[12]  C. Mantzoros,et al.  Obesity and cancer risk: Emerging biological mechanisms and perspectives. , 2019, Metabolism: clinical and experimental.

[13]  S. Vesely,et al.  Paradoxical effects of obesity on T cell function during tumor progression and PD-1 checkpoint blockade , 2018, Nature Medicine.

[14]  Kaushal Parikh,et al.  Use of broad-spectrum antibiotics impacts outcome in patients treated with immune checkpoint inhibitors , 2018, Oncoimmunology.

[15]  M. Mishaeli,et al.  BRAF Mutant Lung Cancer: Programmed Death Ligand 1 Expression, Tumor Mutational Burden, Microsatellite Instability Status, and Response to Immune Check‐Point Inhibitors , 2018, Journal of thoracic oncology : official publication of the International Association for the Study of Lung Cancer.

[16]  W. Willett,et al.  Overall and Central Obesity and Risk of Lung Cancer: A Pooled Analysis , 2018, Journal of the National Cancer Institute.

[17]  Kaushal Parikh,et al.  Peripheral monocytes and neutrophils predict response to immune checkpoint inhibitors in patients with metastatic non-small cell lung cancer , 2018, Cancer Immunology, Immunotherapy.

[18]  L. Zitvogel,et al.  Negative association of antibiotics on clinical activity of immune checkpoint inhibitors in patients with advanced renal cell and non-small-cell lung cancer , 2018, Annals of oncology : official journal of the European Society for Medical Oncology.

[19]  Laurence Zitvogel,et al.  Gut microbiome influences efficacy of PD-1–based immunotherapy against epithelial tumors , 2018, Science.

[20]  Shannon K. Boi,et al.  Obesity alters immune and metabolic profiles: New insight from obese‐resistant mice on high‐fat diet , 2016, Obesity.

[21]  A. Renehan,et al.  The Obesity Paradox in Cancer: a Review , 2016, Current Oncology Reports.

[22]  Y. Abed,et al.  Obesity and inflammation: the linking mechanism and the complications , 2016, Archives of medical science : AMS.

[23]  R. Gomis,et al.  Akkermansia muciniphila inversely correlates with the onset of inflammation, altered adipose tissue metabolism and metabolic disorders during obesity in mice , 2015, Scientific Reports.

[24]  F. Levenez,et al.  Akkermansia muciniphila and improved metabolic health during a dietary intervention in obesity: relationship with gut microbiome richness and ecology , 2015, Gut.

[25]  Marcin Gierach,et al.  Correlation between Body Mass Index and Waist Circumference in Patients with Metabolic Syndrome , 2014, ISRN endocrinology.

[26]  M. Lazar,et al.  The Health Risk of Obesity—Better Metrics Imperative , 2013, Science.

[27]  Isabel Azevedo,et al.  Chronic Inflammation in Obesity and the Metabolic Syndrome , 2010, Mediators of inflammation.

[28]  A. Butler,et al.  Obesity accelerates thymic aging. , 2009, Blood.

[29]  Paul L. Huang A comprehensive definition for metabolic syndrome , 2009, Disease Models & Mechanisms.

[30]  R. Yu,et al.  Visceral Fat Accumulation Induced by a High‐fat Diet Causes the Atrophy of Mesenteric Lymph Nodes in Obese Mice , 2008, Obesity.

[31]  R. D'Agostino,et al.  Metabolic Syndrome as a Precursor of Cardiovascular Disease and Type 2 Diabetes Mellitus , 2005, Circulation.

[32]  M Frost,et al.  Facts and fantasies. , 1978, Nursing times.

[33]  Robert C. Wolpert,et al.  A Review of the , 1985 .