Elsevier

Nutrition

Volume 28, Issue 10, October 2012, Pages 1028-1035
Nutrition

Applied nutritional investigation
Targeting insulin inhibition as a metabolic therapy in advanced cancer: A pilot safety and feasibility dietary trial in 10 patients

https://doi.org/10.1016/j.nut.2012.05.001Get rights and content

Abstract

Objective

Most aggressive cancers demonstrate a positive positron emission tomographic (PET) result using 18F-2-fluoro-2-deoxyglucose (FDG), reflecting a glycolytic phenotype. Inhibiting insulin secretion provides a method, consistent with published mechanisms, for limiting cancer growth.

Methods

Eligible patients with advanced incurable cancers had a positive PET result, an Eastern Cooperative Oncology Group performance status of 0 to 2, normal organ function without diabetes or recent weight loss, and a body mass index of at least 20 kg/m2. Insulin inhibition, effected by a supervised carbohydrate dietary restriction (5% of total kilocalories), was monitored for macronutrient intake, body weight, serum electrolytes, β-hydroxybutyrate, insulin, and insulin-like growth factors-1 and -2. An FDG-PET scan was obtained at study entry and exit.

Results

Ten subjects completed 26 to 28 d of the study diet without associated unsafe adverse effects. Mean caloric intake decreased 35 ± 6% versus baseline, and weight decreased by a median of 4% (range 0.0–6.1%). In nine patients with prior rapid disease progression, five with stable disease or partial remission on PET scan after the diet exhibited a three-fold higher dietary ketosis than those with continued progressive disease (n = 4, P = 0.018). Caloric intake (P = 0.65) and weight loss (P = 0.45) did not differ in those with stable disease or partial remission versus progressive disease. Ketosis was associated inversely with serum insulin levels (P = 0.03).

Conclusion

Preliminary data demonstrate that an insulin-inhibiting diet is safe and feasible in selected patients with advanced cancer. The extent of ketosis, but not calorie deficit or weight loss, correlated with stable disease or partial remission. Further study is needed to assess insulin inhibition as complementary to standard cytotoxic and endocrine therapies.

Introduction

Persistent aerobic glycolysis is a feature of many cancers, although not as universal as originally proposed by Warburg [1]. A glycolytic phenotype nonetheless can be identified in diverse malignancies [2], [3]. Overexpression of the insulin-independent glucose transporter-1 (GLUT-1) [4], [5], [6] and hexokinase [7], [8] facilitates the increased glucose uptake needed to supply the energy needs of these cancers. 18F-2- fluoro-2-deoxyglucose (FDG) undergoes a similar transport and phosphorylation as glucose, its congener. The FDG uptake can be demonstrated on positron emission tomographic (PET) scans of glycolytic cancers, providing a useful tool for the diagnosis, staging, prognosis, and management of numerous aggressive malignancies [9], [10], [11], [12], [13].

The role of insulin in cancer is currently of research interest, and hyperinsulinemia has been described as a risk factor for many cancers [14], [15], [16], [17]. Conversely, we proposed previously that insulin inhibition (INSINH), by altering the metabolic microenvironment, may inhibit many human cancers evolutionarily adapted to a markedly different, specifically hyperinsulinemic, state [18]. We also previously reported on the growth and adenosine triphosphate inhibition in multiple aggressive cancer cell lines when grown in supplemental ketone body medium that are not seen in control fibroblasts [19]. The hypothesis also bears on recent interest in calorie restriction because studies by Kalaany and Sabatini [21] and Sengupta et al. [22], for example, have shown that calorie restriction shares many of the downstream signaling pathways of the insulin receptor.

Mechanistically, the binding of insulin to the insulin receptor activates the mitogen-activated protein (MAP) kinase and phosphatidylinositol-3-kinase pathways in normal cells and different tumor cell lines [22]. Other insulin receptor ligands, including insulin-like growth factor-1 (IGF-1) and IGF-2, share extensive homology and downstream signaling pathways with insulin, but have more potent mitogenic and antiapoptotic effects. In addition to the insulin receptor, IGF-1 receptor, a transmembrane receptor for IGF-1, is upregulated in different human cancers. Ligand binding to the IGF-1 receptor activates its tyrosine kinase, resulting in downstream signaling cascades in the insulin receptor substrate-1(IRS-1)/phosphatidylinositol-3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) and Ras/Raf/mitogen-activated protein kinase/extracellular signal-regulated kinase (ERK) pathways, ultimately promoting proliferation, survival, transformation, metastases, and angiogenesis in many cancers, but especially colorectal and breast cancers. Conversely, decreased insulin secretion induces metabolic and molecular responses, including the inhibition and downregulation of the mammalian target of rapamycin, phosphatidylinositol-3-kinase/Akt, hypoxia-inducible factor (HIF)-1α, fatty acid synthase, and vascular endothelial growth factor (VEGF) and the upregulation of adenosine monophosphate–activated protein kinase (AMPK), all proposed cancer therapy targets [7], [20], [23], [24], [25], [26], [27], [28], [29], [30], [31], [32], [33], [34], [35], [36], [37], [38], [39], [40], [41], [42], [43], [44], [45] of drugs such as rapamycin, wortmannin, bevacizumab, metformin, among many others.

Insulin secretion is inhibited most simply by restricting carbohydrate (CHO) ingestion, thus decreasing the dietary sources of glucose, the principal secretagogue for pancreatic insulin release [46], [47], [48], [49]. The regulation of GLUT-1 translocation by insulin levels has been reported in cancer [50], which can decrease the nutrient supply for glucose-dependent cancers [7], [20], [23], [24], [25], [26], [27], [28], [29], [30], [31], [32], [33], [34], [35], [36], [37], [38], [39], [40], [41], [42], [43], [44], [45]. Ketosis alone and the increased lipolysis that accompany the disinhibition by insulin have been reported to inhibit cancer growth [18], [19], [51], [52], [53], [54], [55], [56], [57], with recent studies demonstrating in vitro [19] and in vivo [54], [58], [59], [60], [61] mechanisms. Further, the adverse effects of CHO-restricted diets have not been demonstrated in normal subjects [62], diabetics [49], [63], or individuals seeking weight loss in studies ranging from 3 mo to 2 y [64], [65], [66], [67], [68] or in patients with cancer over a duration of a 3 mo [69]. Humans with cancer have exhibited a normal nitrogen balance after 1 wk of dietary CHO restriction [52].

Dietary change alone is unlikely to be useful as a cancer therapy, but adding current or developing metabolic, endocrine, and molecular treatments can plausibly increase its effectiveness. Therefore, we initiated a prospective safety and feasibility trial of an INSINH CHO-restricted diet in patients with advanced glucose-dependent PET-FDG–positive cancers. The diets were designed to be eucaloric and weight stable. A change in FDG tumor uptake on a PET scan was chosen as a surrogate marker of a biologic effect [70].

Section snippets

Eligibility criteria

Eligible patients had incurable, advanced cancer with FDG-avid tumors detected by PET scanning, with progressive disease after at least two conventional anticancer treatments. The exclusion criteria included a body mass index lower than 20 kg/m2, a weight loss exceeding 5% of body weight within 3 mo of enrollment, a history of diabetes on hypoglycemic medications, intestinal obstruction, and abnormal liver function (increase in total or direct bilirubin to 1.1 or 0.3 mg/dL, respectively, and

Patient characteristics, dietary adherence, and adverse effects

Twelve patients with advanced cancer were recruited. For reasons unrelated to the intervention, two patients discontinued the study in less than 14 d and therefore were not evaluated. Of these two patients, one withdrew because of symptomatic chest wall disease on day 2 of the diet, requiring hospitalization and chemotherapy; the second patient withdrew after 1 wk because of clinical depression. The remaining 10 patients were included in the results; of these, five patients completed all 28 d

Discussion

The metabolic effects caused by the insulin inhibitory response to CHO restriction may result in disease stabilization in selected cancer types. Cancers cultured in glucose medium in vitro have been inhibited by supplemental ketone bodies [19], [51], [53] and the inhibition of tumor growth in a xenograft model has been associated with ketosis [56] and in non-ketotic rodent models limiting CHO ingestion [53], [54], [58], [59]. In human case reports, glioblastoma demonstrated partial remission on

Conclusion

Insulin inhibition effected by dietary CHO restriction was found safe and feasible in 10 patients with advanced cancer. The three-fold higher ketosis, demonstrated in patients with SD or PR compared with those with continued PD, must be interpreted cautiously in this small pilot study.

Acknowledgments

We are grateful for the consistent support of Dr. Shalom Kalnicki.

References (96)

  • K.C. Fearon et al.

    Cancer cachexia: influence of systemic ketosis on substrate levels and nitrogen metabolism

    Am J Clin Nutr

    (1988)
  • J.C. Mavropoulos et al.

    Is there a role for a low-carbohydrate ketogenic diet in the management of prostate cancer?

    Urology

    (2006)
  • M.J. Sharman et al.

    A ketogenic diet favorably affects serum biomarkers for cardiovascular disease in normal-weight men

    J Nutr

    (2002)
  • H. Young et al.

    Measurement of clinical and subclinical tumour response using [18F]-fluorodeoxyglucose and positron emission tomography: review and 1999 EORTC recommendations. European Organization for Research and Treatment of Cancer (EORTC) PET Study Group

    Eur J Cancer

    (1999)
  • P. Carde et al.

    Early response to chemotherapy: a surrogate for final outcome of Hodgkin's disease patients that should influence initial treatment length and intensity?

    Ann Oncol

    (2002)
  • W. Romer et al.

    Positron emission tomography in non-Hodgkin's lymphoma: assessment of chemotherapy with fluorodeoxyglucose

    Blood

    (1998)
  • K. Spaepen et al.

    Early restaging positron emission tomography with (18)F-fluorodeoxyglucose predicts outcome in patients with aggressive non-Hodgkin's lymphoma

    Ann Oncol

    (2002)
  • A.M. Johnstone et al.

    Effects of a high-protein ketogenic diet on hunger, appetite, and weight loss in obese men feeding ad libitum

    Am J Clin Nutr

    (2008)
  • V.D. Longo et al.

    Calorie restriction and cancer prevention: metabolic and molecular mechanisms

    Trends Pharmacol Sci

    (2010)
  • J.B. Johnson et al.

    Pretreatment with alternate day modified fast will permit higher dose and frequency of cancer chemotherapy and better cure rates

    Med Hypotheses

    (2009)
  • O. Warburg

    On the origin of cancer cells

    Science

    (1956)
  • R.J. Gillies et al.

    Causes and consequences of increased glucose metabolism of cancers

    J Nucl Med

    (2008)
  • M.G. Vander Heiden et al.

    Understanding the Warburg effect: the metabolic requirements of cell proliferation

    Science

    (2009)
  • P.J. Hoskin et al.

    GLUT1 and CAIX as intrinsic markers of hypoxia in bladder cancer: relationship with vascularity and proliferation as predictors of outcome of ARCON

    Br J Cancer

    (2003)
  • D.W. Lambert et al.

    Molecular changes in the expression of human colonic nutrient transporters during the transition from normality to malignancy

    Br J Cancer

    (2002)
  • H. Abdel-Nabi et al.

    Staging of primary colorectal carcinomas with fluorine-18 fluorodeoxyglucose whole-body PET: correlation with histopathologic and CT findings

    Radiology

    (1998)
  • S. Bang et al.

    The clinical usefulness of 18-fluorodeoxyglucose positron emission tomography in the differential diagnosis, staging, and response evaluation after concurrent chemoradiotherapy for pancreatic cancer

    J Clin Gastroenterol

    (2006)
  • M. Findlay et al.

    Noninvasive monitoring of tumor metabolism using fluorodeoxyglucose and positron emission tomography in colorectal cancer liver metastases: correlation with tumor response to fluorouracil

    J Clin Oncol

    (1996)
  • W.A. Weber et al.

    Monitoring chemotherapy and radiotherapy of solid tumors

    Eur J Nucl Med Mol Imaging

    (2006)
  • M.J. Gunter et al.

    A prospective evaluation of insulin and insulin-like growth factor-I as risk factors for endometrial cancer

    Cancer Epidemiol Biomarkers Prev

    (2008)
  • M.J. Gunter et al.

    Insulin, insulin-like growth factor-I, endogenous estradiol, and risk of colorectal cancer in postmenopausal women

    Cancer Res

    (2008)
  • D. Lann et al.

    The role of endocrine insulin-like growth factor-I and insulin in breast cancer

    J Mammary Gland Biol Neoplasia

    (2008)
  • D. Albanes et al.

    Serum insulin, glucose, indices of insulin resistance, and risk of prostate cancer

    J Natl Cancer Inst

    (2009)
  • E. Fine et al.

    Carbohydrate restriction in patients with advanced cancer: a protocol to assess safety and feasibility with an accompanying hypothesis

    Community Oncol

    (2008)
  • E.J. Fine et al.

    Acetoacetate reduces growth and ATP concentration in cancer cell lines which over-express uncoupling protein 2

    Cancer Cell Int

    (2009)
  • N.Y. Kalaany et al.

    Tumours with PI3K activation are resistant to dietary restriction

    Nature

    (2009)
  • S. Sengupta et al.

    mTORC1 controls fasting-induced ketogenesis and its modulation by ageing

    Nature

    (2010)
  • D.B. Shackelford et al.

    The LKB1-AMPK pathway: metabolism and growth control in tumour suppression

    Nat Rev Cancer

    (2009)
  • A.S. Fung et al.

    Concurrent and sequential administration of chemotherapy and the mammalian target of rapamycin inhibitor temsirolimus in human cancer cells and xenografts

    Clin Cancer Res

    (2009)
  • J.C. Yao et al.

    Daily oral everolimus activity in patients with metastatic pancreatic neuroendocrine tumors after failure of cytotoxic chemotherapy: a phase II trial

    J Clin Oncol

    (2010)
  • T. David-Pfeuty et al.

    Targeting the cell cycle and the PI3K pathway: a possible universal strategy to reactivate innate tumor suppressor programmes in cancer cells

    Int J Oncol

    (2010)
  • M. Lopez-Fauqued et al.

    The dual PI3K/mTOR inhibitor PI-103 promotes immunosuppression, in vivo tumor growth and increases survival of sorafenib-treated melanoma cells

    Int J Cancer

    (2010)
  • L.A. Cantrell et al.

    Metformin is a potent inhibitor of endometrial cancer cell proliferation—implications for a novel treatment strategy

    Gynecol Oncol

    (2009)
  • J.N. Lin et al.

    Resveratrol modulates tumor cell proliferation and protein translation via SIRT1-dependent AMPK activation

    J Agric Food Chem

    (2010)
  • Z. Luo et al.

    AMPK as a metabolic tumor suppressor: control of metabolism and cell growth

    Future Oncol

    (2010)
  • M.Y. Koh et al.

    Inhibiting the hypoxia response for cancer therapy: the new kid on the block

    Clin Cancer Res

    (2009)
  • R.J. Leeman-Neill et al.

    Guggulsterone enhances head and neck cancer therapies via inhibition of signal transducer and activator of transcription-3

    Carcinogenesis

    (2009)
  • X. Sun et al.

    Antisense hypoxia-inducible factor-1alpha augments transcatheter arterial embolization in the treatment of hepatocellular carcinomas in rats

    Hum Gene Ther

    (2009)
  • Cited by (142)

    • Dietary approaches for exploiting metabolic vulnerabilities in cancer

      2024, Biochimica et Biophysica Acta - Reviews on Cancer
    • Cancer

      2023, Ketogenic: The Science of Therapeutic Carbohydrate Restriction in Human Health
    • Ketogenic diet and cancer

      2022, Medecine des Maladies Metaboliques
    View all citing articles on Scopus

    This work was supported by the State University of New York Research Foundation and the Robert and Veronica Atkins Foundation. This publication also was supported in part by CTSA grants UL1RR025750, KL2RR025749, and TL1RR025748 from the National Center for Research Resources, a component of the National Institutes of Health, and the National Institutes of Health Roadmap for Medical Research.

    View full text