Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Neil M. Iyengar is active.

Publication


Featured researches published by Neil M. Iyengar.


Annual Review of Medicine | 2015

Obesity and Cancer: Local and Systemic Mechanisms

Neil M. Iyengar; Clifford A. Hudis; Andrew J. Dannenberg

Obesity is a leading modifiable risk factor for the development of several epithelial malignancies. In addition to increasing risk, obesity also confers worse prognosis for many cancers. Obesity represents an overall state of energy imbalance frequently associated with systemic effects including insulin resistance, altered hormone signaling, and high circulating levels of proinflammatory mediators. In addition to its systemic effects, obesity causes subclinical white adipose inflammation including increased tissue levels of proinflammatory mediators. Both local and systemic effects are likely to contribute to the development and progression of cancer. An understanding of the interplay between local and systemic alterations involved in the obesity-cancer link provides the basis for developing interventions aimed at mitigating the protumorigenic effects.


Clinical Cancer Research | 2016

Systemic Correlates of White Adipose Tissue Inflammation in Early-Stage Breast Cancer

Neil M. Iyengar; Xi Kathy Zhou; Ayca Gucalp; Patrick G. Morris; Louise R. Howe; Dilip Giri; Monica Morrow; Hanhan Wang; Michael Pollak; Lee W. Jones; Clifford A. Hudis; Andrew J. Dannenberg

Purpose: Obesity, insulin resistance, and elevated levels of circulating proinflammatory mediators are associated with poorer prognosis in early-stage breast cancer. To investigate whether white adipose tissue (WAT) inflammation represents a potential unifying mechanism, we examined the relationship between breast WAT inflammation and the metabolic syndrome and its prognostic importance. Experimental Design: WAT inflammation was defined by the presence of dead/dying adipocytes surrounded by macrophages forming crown-like structures (CLS) of the breast. Two independent groups were examined in cross-sectional (cohort 1) and retrospective (cohort 2) studies. Cohort 1 included 100 women undergoing mastectomy for breast cancer risk reduction (n = 10) or treatment (n = 90). Metabolic syndrome–associated circulating factors were compared by CLS-B status. The association between CLS of the breast and the metabolic syndrome was validated in cohort 2, which included 127 women who developed metastatic breast cancer. Distant recurrence-free survival (dRFS) was compared by CLS-B status. Results: In cohorts 1 and 2, breast WAT inflammation was detected in 52 of 100 (52%) and 52 of 127 (41%) patients, respectively. Patients with breast WAT inflammation had elevated insulin, glucose, leptin, triglycerides, C-reactive protein, and IL6 and lower high-density lipoprotein cholesterol and adiponectin (P < 0.05) in cohort 1. In cohort 2, breast WAT inflammation was associated with hyperlipidemia, hypertension, and diabetes (P < 0.05). Compared with patients without breast WAT inflammation, the adjusted HR for dRFS was 1.83 (95% CI, 1.07–3.13) for patients with inflammation. Conclusions: WAT inflammation, a clinically occult process, helps to explain the relationship between metabolic syndrome and worse breast cancer prognosis. Clin Cancer Res; 22(9); 2283–9. ©2015 AACR.


Journal of Clinical Oncology | 2016

Obesity and Cancer Mechanisms: Tumor Microenvironment and Inflammation.

Neil M. Iyengar; Ayca Gucalp; Andrew J. Dannenberg; Clifford A. Hudis

Purpose There is growing evidence that inflammation is a central and reversible mechanism through which obesity promotes cancer risk and progression. Methods We review recent findings regarding obesity-associated alterations in the microenvironment and the local and systemic mechanisms through which these changes support tumor growth. Results Locally, hyperadiposity is associated with altered adipose tissue function, adipocyte death, and chronic low-grade inflammation. Most individuals who are obese harbor inflamed adipose tissue, which resembles chronically injured tissue, with immune cell infiltration and remodeling. Within this distinctly altered local environment, several pathophysiologic changes are found that may promote breast and other cancers. Consistently, adipose tissue inflammation is associated with a worse prognosis in patients with breast and tongue cancers. Systemically, the metabolic syndrome, including dyslipidemia and insulin resistance, occurs in the setting of adipose inflammation and operates in concert with local mechanisms to sustain the inflamed microenvironment and promote tumor growth. Importantly, adipose inflammation and its protumor consequences can be found in some individuals who are not considered to be obese or overweight by body mass index. Conclusion The tumor-promoting effects of obesity occur at the local level via adipose inflammation and associated alterations in the microenvironment, as well as systemically via circulating metabolic and inflammatory mediators associated with adipose inflammation. Accurately characterizing the obese state and identifying patients at increased risk for cancer development and progression will likely require more precise assessments than body mass index alone. Biomarkers of adipose tissue inflammation would help to identify high-risk populations. Moreover, adipose inflammation is a reversible process and represents a novel therapeutic target that warrants further study to break the obesity-cancer link.


Journal of Clinical Oncology | 2015

Phase II Study of Paclitaxel Given Once per Week Along With Trastuzumab and Pertuzumab in Patients With Human Epidermal Growth Factor Receptor 2–Positive Metastatic Breast Cancer

Chau Dang; Neil M. Iyengar; Fm Datko; Gabriella D'Andrea; Maria Theodoulou; Maura N. Dickler; Shari Goldfarb; Diana Lake; Julie Fasano; Monica Fornier; Theresa Gilewski; Shanu Modi; Devika Gajria; Mary Ellen Moynahan; Nicola Hamilton; Sujata Patil; Maxine S. Jochelson; Larry Norton; José Baselga; Clifford A. Hudis

PURPOSE The CLEOPATRA (Clinical Evaluation of Trastuzumab and Pertuzumab) study demonstrated superior progression-free survival (PFS) and overall survival when pertuzumab was added to trastuzumab and docetaxel. Paclitaxel given once per week is effective and less toxic than docetaxel. We performed a phase II study to evaluate the efficacy and safety of pertuzumab and trastuzumab with paclitaxel given once per week. PATIENTS AND METHODS Patients with metastatic human epidermal growth factor receptor 2-positive breast cancer with zero to one prior therapy were enrolled. Treatment consisted of paclitaxel 80 mg/m(2) once per week plus trastuzumab (8 mg/kg loading dose → 6 mg/kg) once every 3 weeks plus pertuzumab (840 mg loading dose → 420 mg) once every 3 weeks, all given intravenously. The primary end point was 6-month PFS assessed by Kaplan-Meier methods. RESULTS From January 2011 to December 2013, we enrolled 69 patients: 51 (74%) and 18 (26%) treated in first- and second-line metastatic settings, respectively. At a median follow-up of 21 months (range, 3 to 38 months), 6-month PFS was 86% (95% CI, 75% to 92%). The median PFS was 19.5 months (95% CI, 14 to 26 months) overall. PFS was 24.2 months (95% CI, 14 months to not reached [NR]) and 16.4 months (95% CI, 8.5 months to NR) for those without and with prior treatment, respectively. At 1 year, Kaplan-Meier PFS was 70% (95% CI, 56% to 79%) overall, 71% (95% CI, 55% to 82%) for those without prior therapy, and 66% (95% CI, 40% to 83%) for those with prior therapy. Treatment was well-tolerated; there was no febrile neutropenia or symptomatic left ventricular systolic dysfunction. CONCLUSION Paclitaxel given once per week with trastuzumab and pertuzumab is highly active and well tolerated and seems to be an effective alternative to docetaxel-based combination therapy.


Cancer Prevention Research | 2015

Menopause Is a Determinant of Breast Adipose Inflammation

Neil M. Iyengar; Patrick G. Morris; Xi Kathy Zhou; Ayca Gucalp; Dilip Giri; Michael D. Harbus; Domenick J. Falcone; Margaret Krasne; Linda T. Vahdat; Kotha Subbaramaiah; Monica Morrow; Clifford A. Hudis; Andrew J. Dannenberg

Chronic inflammation is recognized as a risk factor for the development of several malignancies. Local white adipose tissue (WAT) inflammation, defined by the presence of dead or dying adipocytes encircled by macrophages that form crown-like structures (CLS), occurs in the breasts (CLS-B) of most overweight and obese women. Previously, we showed that the presence of CLS-B is associated with elevated tissue levels of proinflammatory mediators and aromatase, the rate-limiting enzyme for estrogen biosynthesis. The associated increased levels of aromatase in the breast provide a plausible mechanistic link between WAT inflammation and estrogen-dependent breast cancers. Thus, breast WAT inflammation could be relevant for explaining the high incidence of estrogen-dependent tumors with aging despite diminished circulating estrogen levels after menopause. To explore this possibility, we determined whether menopause in addition to body mass index (BMI) is associated with breast WAT inflammation among 237 prospectively enrolled women. The presence of CLS-B and its severity (CLS-B/cm2) as indicators of WAT inflammation correlated with menopausal status (P = 0.008 and P < 0.001) and BMI (P < 0.001 for both). In multivariable analyses adjusted for BMI, the postmenopausal state was independently associated with the presence (P = 0.03) and severity of breast WAT inflammation (P = 0.01). Mean adipocyte size increased in association with CLS-B (P < 0.001). Our findings demonstrate that breast WAT inflammation, which is associated with elevated aromatase levels, is increased in association with the postmenopausal state independent of BMI. Breast WAT inflammation, a process that can potentially be targeted, may help to explain the high incidence of estrogen-dependent tumors in postmenopausal women. Cancer Prev Res; 8(5); 349–58. ©2015 AACR.


Cancer | 2014

Impact of obesity on the survival of patients with early-stage squamous cell carcinoma of the oral tongue.

Neil M. Iyengar; Amit Kochhar; Patrick G. Morris; Luc G. T. Morris; Xi K. Zhou; Ronald Ghossein; Alejandro Pino; Matthew G. Fury; David G. Pfister; Snehal G. Patel; Jay O. Boyle; Clifford A. Hudis; Andrew J. Dannenberg

Although obesity increases risk and negatively affects survival for many malignancies, the prognostic implications in squamous cell carcinoma (SCC) of the oral tongue, a disease often associated with prediagnosis weight loss, are unknown.


Cancer Prevention Research | 2017

Metabolic obesity, adipose inflammation and elevated breast aromatase in women with normal body mass index

Neil M. Iyengar; Kristy A. Brown; Xi Kathy Zhou; Ayca Gucalp; Kotha Subbaramaiah; Dilip Giri; Heba Zahid; Priya Bhardwaj; Nils K. Wendel; Domenick J. Falcone; Hanhan Wang; Samantha Williams; Michael Pollak; Monica Morrow; Clifford A. Hudis; Andrew J. Dannenberg

Obesity is associated with breast white adipose tissue (WAT) inflammation, elevated levels of the estrogen biosynthetic enzyme, aromatase, and systemic changes that have been linked to the pathogenesis of breast cancer. Here, we determined whether metabolic obesity, including changes in breast biology and systemic effects, occurs in a subset of women with normal body mass index (BMI). Breast WAT and fasting blood were collected from 72 women with normal BMI (<25 kg/m2) undergoing mastectomy for breast cancer risk reduction or treatment. WAT inflammation was defined by the presence of crown-like structures of the breast (CLS-B) which are composed of dead or dying adipocytes surrounded by macrophages. Severity of inflammation was measured as CLS-B/cm2. The primary objective was to determine whether breast WAT inflammation is associated with aromatase expression and activity. Secondary objectives included assessment of circulating factors and breast adipocyte size. Breast WAT inflammation was present in 39% of women. Median BMI was 23.0 kg/m2 (range, 18.4–24.9 kg/m2) in women with breast WAT inflammation versus 21.8 kg/m2 (range, 17.3–24.6 kg/m2) in those without inflammation (P = 0.04). Breast WAT inflammation was associated with elevated aromatase expression and activity, which increased with severity of inflammation (P < 0.05). Breast WAT inflammation correlated with larger adipocytes (P = 0.01) and higher circulating levels of C-reactive protein, leptin, insulin, and triglycerides (P ≤ 0.05). A subclinical inflammatory state associated with elevated aromatase in the breast, adipocyte hypertrophy, and systemic metabolic dysfunction occurs in some normal BMI women and may contribute to the pathogenesis of breast cancer. Cancer Prev Res; 10(4); 235–43. ©2017 AACR. See related article by Berger, p. 223–25.


Archive | 2013

Obesity, Inflammation, and Breast Cancer

Neil M. Iyengar; Patrick G. Morris; Clifford A. Hudis; Andrew J. Dannenberg

Obesity, which is rising in incidence worldwide, is important with regard to the treatment of breast cancer, disease progression, and carcinogenesis. Obesity is a risk factor for the development of hormone receptor-positive breast cancer in postmenopausal women and is associated with reduced benefits from treatment. Furthermore, irrespective of breast cancer subtype, obesity is associated with worse outcomes after diagnosis. There is increasing evidence of specific biological underpinnings for these observations, including higher circulating estrogen levels, insulin resistance, altered levels of adipokines, and the consequences of chronic in-breast inflammation. Increasing adiposity also has important implications for local therapy including surgery and radiotherapy. This chapter reviews the complex interactions between obesity and breast cancer.


Current Breast Cancer Reports | 2013

Omega-3 Fatty Acids for Prevention of Breast Cancer: an Update and the State of the Science

Neil M. Iyengar; Clifford A. Hudis; Ayca Gucalp

The quantity and makeup of dietary fat intake are known to affect human health. A variety of purported health benefits, including cancer prevention, have focused increased attention on use of omega-3 (ω-3) polyunsaturated fatty acid (PUFA) supplements. Preclinical evidence has been encouraging, and recent studies have increased our understanding of mechanisms by means of which ω-3 PUFAs may protect against breast cancer. However, epidemiological studies have yielded mixed results. Recent population studies have attempted to determine factors, for example total fat intake and the ratio of ω-3 to ω-6 PUFA intake, that may affect the action of ω-3 PUFAs. Several clinical trials, some currently ongoing, are investigating strategies to favorably alter endogenous fatty acid profiles in an attempt to develop clinically feasible prevention methods. Identification of well-defined subpopulations who are most likely to benefit from a targeted prevention approach will probably be crucial to this effort.


Cancer | 2013

Long‐term cardiac safety and outcomes of dose‐dense doxorubicin and cyclophosphamide followed by paclitaxel and trastuzumab with and without lapatinib in patients with early breast cancer

Patrick G. Morris; Neil M. Iyengar; Sujata Patil; Carol Chen; Alyson Abbruzzi; Robert Lehman; Richard M. Steingart; Kevin C. Oeffinger; Nan Lin; Beverley Moy; Steven E. Come; Larry Norton; Clifford A. Hudis; Chau T. Dang

The authors have previously reported 2 consecutive phase 2 trials in patients with early breast cancer that overexpresses human epidermal growth factor receptor 2 (HER2) to assess the feasibility of incorporating anti‐HER2 therapies into dose‐dense (dd) chemotherapy regimens. The incidence of congestive heart failure (CHF) at a median follow‐up of 2 years was 1.4% and 3.2%, respectively.

Collaboration


Dive into the Neil M. Iyengar's collaboration.

Top Co-Authors

Avatar

Clifford A. Hudis

American Society of Clinical Oncology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ayca Gucalp

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Dilip Giri

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chau T. Dang

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Larry Norton

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Sujata Patil

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

José Baselga

Memorial Sloan Kettering Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge