Mocht u de artikelen op kanker-actueel waarderen dan wilt u ons misschien ondersteunen met een donatie op 

RABO 37.29.31.138 t.n.v. Stichting Gezondheid Actueel in Terneuzen.
Onze IBANcode is NL79 RABO 0372 9311 38
BIC/SWIFTCODE RABONNL2U

5 april 2019: lees ook dit artikel: 

https://kanker-actueel.nl/vrouw-van-29-jaar-met-triple-negatieve-borstkanker-stadium-iv-komt-in-complete-remissie-door-aanvullend-op-chemo-persoonlijke-voedingsadviezen-kytogeen-dieet-hyperthermie-en-hyperbare-zuurstoftherapie.html

3 april 2019: Bron: International Journal of Hyperthermie

Patiënten met gevorderde niet-kleincellige longkanker (stadium IV) blijken veel baat te kunnen hebben bij aanvullend op chemo (carboplatin / paclitaxel ) een combinatie van hyperthermie, een persoonlijk voedingsprogramma op basis van het kytogene dieet en hyperbare zuurstoftherapie. Maar liefst 61,4 procent van de 44 deelnemende patienten reageerde met een complete remissie of een gedeeltelijke remissie (50 procent of meer). Slechts 22 procent had progressieve ziekte dus bij die patiënten had de aanpak geen succes. 15 patienten overleden voordat de 5-jarige studie eindigde maar 29 patienten waren nog in leven waarvan een aantal met progressievrije ziekte.  

Meadiane overall overleving (OS)  was 42.9 maanden (95% CI: 34.0–51.8) en progressievrije overleving (PFS) 41.0 maanden (95% CI: 31.1–50.9) wat extreem lang is voor deze groep van longkankerpatienten. De studie duurde van maart 2010 tot juni 2015. Het studierapport werd 1 april 2019 gepubliceerd.

Bijwerkingenprofielen per individuele patiënt waren goed te controleren aldus de onderzoekers.

Ik moet wel zeggen dat alle patienten heel intensief werden behandeld want om naast chemo ook hyperthermie te geven is heel gemakkelijk te doen. Maar voor elke patient werd ook een voedingsadvies samengesteld gegeven op basis van haar / zijn persoonlijke lichamelijke gestelheid op basis van het kytogeen dieet. En daarnaast kregen alle patienten ook nog eens sessies met hyperbare zuurstoftherapie. Maar het blijkt dus zeer de moeite waard te zijn gezien de mediane overall overleving van voor een aantal patienten langer dan 5 jaar. Zelfs uitzaaiingen in de hersenen waren geen contra indicatie, in tegendeel sommige patienten leven al langer dan 5 jaar daarmee. Wel was het zo hoe slechter de lichamelijke gestgeldhied hoe minder de resultaten. Zie Figure 3

Deze opmerkelijke studieresultaten (mediane overall overleving was zelfs beter dan voor immuuntherapie met atezolizumab bv.) werden afgelopen week gepresenteerd in het International Journal of Hyperthermie.

Hier de resultaten zoals gepresenteerd in het volledige studierapport: 

Feasibility study of metabolically supported chemotherapy with weekly carboplatin/paclitaxel combined with ketogenic diet, hyperthermia and hyperbaric oxygen therapy in metastatic non-small cell lung cancer dat gratis is in te zien inclusief grafieken van de karakteristieken van de patienten.

Survival estimates

At the end of 8 cycles of treatment, 42 out of 44 patients (95.4%) were alive and had completed 8 cycles of treatment. Two patients died after receiving 3 cycles of treatment. At the termination of follow-up (15 January 2016), 29 patients were alive (65.9%). Mean overall survival (OS) was 42.9 months (95% CI: 34.0–51.8). Corresponding figure for progression-free survival (PFS) was 41.0 months (95% CI: 31.1–50.9).

Table 3 shows mean survival rates (OS and PFS) by patient characteristics. A higher ECOG status (ECOG ≥2) was associated with worse OS (33.0 vs. 63.9 months, p = .009) and PFS (29.4 vs. 63.4 months, p = .009). On the other hand, age, histology, presence of brain metastasis and smoking had no effect on survival outcome. Figure 3 shows Kaplan–Meier curves for OS. In addition, a subgroup analysis of patients with available ALK fusion data (n = 29) showed numerically better overall survival (61.2 vs. 34.9 months) and progression-free survival rates (61.1 vs. 30.1 months) for patients with fusion (n = 5) than patients without fusion (n = 24); however, the difference did not reach statistical significance (p = .103 and .085, respectively).

Figure 3. Kaplan–Meier curves for overall survival: (A) all patients; and patients stratified by ECOG performance status (B), age (C), and presence of brain metastasis (D). p values are calculated with Log-rank test. A higher ECOG status (ECOG ≥2) was associated with worse overall survival (Panel B, p = .009). However, older age (Panel C) and the presence of brain metastasis (Panel D) did not have an effect on overall survival outcome (p > .05 for both).

Hier het abstract van deze studie:

Feasibility study of metabolically supported chemotherapy with weekly carboplatin/paclitaxel combined with ketogenic diet, hyperthermia and hyperbaric oxygen therapy in metastatic non-small cell lung cancer

Received 10 Sep 2018, Accepted 27 Feb 2019, Published online: 01 Apr 2019

Background: Previous evidence suggests that metabolically supported chemotherapy (MSCT), ketogenic diet, hyperthermia and hyperbaric oxygen therapy (HBOT) could all target vulnerabilities of cancer cells. This study aimed to evaluate the efficacy and the tolerability of this combination therapy in the treatment of stage IV non-small cell lung cancer (NSCLC).

Methods: Forty-four NSCLC patients with distant metastasis that received MSCT (administration of chemotherapy regimen following induced hypoglycemia) plus ketogenic diet, hyperthermia and HBOT combination were included in this retrospective study. Survival and treatment response rates as well as toxicities were evaluated.

Results: Overall response rate (ORR, complete response plus partial response) was 61.4%; whereas, 15.9% and 22.7% of patients had stable disease (SD) and progressive disease (PD), respectively. Mean overall survival (OS) and progression-free survival (PFS) was 42.9 months (95% CI: 34.0–51.8) and 41.0 months (95% CI: 31.1–50.9), respectively. A higher Eastern Cooperative Oncology Group (ECOG) performance status (ECOG ≥2) was associated with worse OS and PFS. Patients received chemotherapy cycles with acceptable toxicity and adverse events. No problems were encountered due to fasting, hypoglycemia, ketogenic diet, hyperthermia or hyperbaric oxygen therapy.

Conclusions: Findings of this study suggest that MSCT combined with other modalities targeting multiple pathways and cellular vulnerabilities may bring about remarkable improvements in survival outcomes and treatment response rates in metastatic NSCLC, without additional safety concerns. Large comparative studies are warranted to draw robust conclusions.

Referentielijst:

Disclosure statement

No potential conflict of interest was reported by the authors.

References

  • Brambilla E, Travis WD. Lung cancer. In: Stewart BW, Wild CP, editors. World cancer report. Lyon: WHO; 2014:350361.  
  • Torre LA, Bray F, Siegel RL, et al. Global cancer statistics, 2012. CA Cancer J Clin. 2015;65:87108. , 
  • Glassberg AB, Cornett P. Lung: non-small cell. In: Dollinger M, Rosenbaum EH, Cable G, editors. Everyone's guide to cancer therapy. Kansas City, MO: Somerville House Books Limited; 1994. p. 469475.  
  • Molina JR, Yang P, Cassivi SD, et al. Non-small cell lung cancer: epidemiology, risk factors, treatment, and survivorship. Mayo Clin Proc. 2008;83:584594. , 
  • Ramalingam S, Belani C. Systemic chemotherapy for advanced non-small cell lung cancer: recent advances and future directions. Oncologist. 2008;13:513. , 
  • Lilenbaum RC. Overview of the initial treatment of advanced non-small cell lung cancer. In: West HJ, editor. UpToDate. 2019. Topic 4607, Version 60.0.  
  • Soejima K, Naoki K, Ishioka K, et al. A phase II study of biweekly paclitaxel and carboplatin in elderly patients with advanced non-small cell lung cancer. Cancer Chemother Pharmacol. 2015;75:513519. , 
  • Reck M, Popat S, Reinmuth N, et al. Metastatic non-small-cell lung cancer (NSCLC): ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2014;25:iii27iii39. , 
  • Kelly K, Crowley J, Bunn PA, Jr, et al. Randomized phase III trial of paclitaxel plus carboplatin versus vinorelbine plus cisplatin in the treatment of patients with advanced non–small-cell lung cancer: a Southwest Oncology Group trial. JCO. 2001;19:32103218. , 
  • Socinski MA, Jotte RM, Cappuzzo F, et al. Atezolizumab for first-line treatment of metastatic nonsquamous NSCLC. N Engl J Med. 2018;378:22882301. , 
  • Schulze AB, Schmidt LH. PD-1 targeted Immunotherapy as first-line therapy for advanced non-small-cell lung cancer patients. J Thorac Dis. 2017;9:E384E386. , 
  • Borghaei H, Hellmann MD, Paz-Ares LG, et al. Nivolumab (Nivo) + platinum-doublet chemotherapy (Chemo) vs chemo as first-line (1L) treatment (Tx) for advanced non-small cell lung cancer (NSCLC) with <1% tumor PD-L1 expression: Results from CheckMate 227. JCO. 2018;36:90019001. , 
  • Lopes G, Wu Y-L, Kudaba I, et al. Pembrolizumab (pembro) versus platinum-based chemotherapy (chemo) as first-line therapy for advanced/metastatic NSCLC with a PD-L1 tumor proportion score (TPS) ≥ 1%: Open-label, phase 3 KEYNOTE-042 study. JCO. 2018;36:LBA4LBA4. , 
  • Carbone DP, Reck M, Paz-Ares L, et al. First-line nivolumab in stage IV or recurrent non-small-cell lung cancer. N Engl J Med. 2017;376:24152426. , 
  • Schiller JH, Harrington D, Belani CP, et al. Comparison of four chemotherapy regimens for advanced non-small-cell lung cancer. N Engl J Med. 2002;346:9298. , 
  • Ichiki M, Gohara R, Fujiki R, et al. Phase I and pharmacokinetic study of carboplatin and paclitaxel with a biweekly schedule in patients with advanced non-small-cell lung cancer. Cancer Chemother Pharmacol. 2003;52:6772. , 
  • Maemondo M, Inoue A, Sugawara S, et al. Randomized phase II trial comparing carboplatin plus weekly paclitaxel and docetaxel alone in elderly patients with advanced non-small cell lung cancer: north japan lung cancer group trial 0801. Oncologist. 2014;19:352353. , 
  • Quoix E, Zalcman G, Oster JP, et al. Carboplatin and weekly paclitaxel doublet chemotherapy compared with monotherapy in elderly patients with advanced non-small-cell lung cancer: IFCT-0501 randomised, phase 3 trial. Lancet. 2011;378:10791088. , 
  • Volk V, Cathomas R, Mark M, et al. Weekly carboplatin in combination with weekly paclitaxel in the treatment of metastatic non-small cell lung cancer: a single center 10-year experience. Support Care Cancer. 2016;24:21192128. , 
  • Liberti MV, Locasale JW. The Warburg effect: how does it benefit cancer cells? Trends Biochem Sci. 2016;41:211218. , 
  • Warburg OK. Uber den Stoffwechsel der Carcinomzelle [About the metabolism of the carcinoma cell]. Biochem Z. 1924;152:309344.  
  • Warburg O. On the origin of cancer cells. Science. 1956;123:309314. , 
  • Seyfried TN, Shelton LM. Cancer as a metabolic disease. Nutr Metab (Lond). 2010;7:7. , 
  • Iyikesici MS, Slocum A, Turkmen E, et al. Long-term outcomes of the treatment of unresectable (Stage III - IV) ductal pancreatic adenocarcinoma using metabolically supported chemotherapy (MSCT): a retrospective study. JOP. 2015;17:3641. , 
  • Iyikesici MS, Slocum A, Turkmen E, et al. Complete response of locally advanced (stage III) rectal cancer to metabolically supported chemoradiotherapy with hyperthermia. Int J Cancer Res Mol Mech. 2016;2:14.  
  • Iyikesici MS, Slocum AK, Slocum A, et al. Efficacy of metabolically supported chemotherapy combined with ketogenic diet, hyperthermia, and hyperbaric oxygen therapy for stage IV triple-negative breast cancer. Cureus. 2017;9:e1445. , 
  • Ayre SG, Garcia y, Bellon DP, Garcia DP. Jr. Insulin, chemotherapy, and the mechanisms of malignancy: the design and the demise of cancer. Med Hypotheses. 2000;55:330334. , 
  • Seyfried TN, Flores R, Poff AM, et al. Metabolic therapy: a new paradigm for managing malignant brain cancer. Cancer Lett. 2015;356:289300. , 
  • Stafford P, Abdelwahab MG, Kim DY, et al. The ketogenic diet reverses gene expression patterns and reduces reactive oxygen species levels when used as an adjuvant therapy for glioma. Nutr Metab (Lond). 2010;7:74. , 
  • Masko EM, Thomas JA, 2nd, Antonelli JA, et al. Low-carbohydrate diets and prostate cancer: how low is "low enough"? Cancer Prev Res (Phila). 2010;3:11241131. , 
  • Zuccoli G, Marcello N, Pisanello A, et al. Metabolic management of glioblastoma multiforme using standard therapy together with a restricted ketogenic diet: case report. Nutr Metab (Lond). 2010;7:33. , 
  • Zhou W, Mukherjee P, Kiebish MA, et al. The calorically restricted ketogenic diet, an effective alternative therapy for malignant brain cancer. Nutr Metab (Lond). 2007;4:5. , 
  • Poff AM, Ari C, Seyfried TN, et al. The ketogenic diet and hyperbaric oxygen therapy prolong survival in mice with systemic metastatic cancer. PLoS One. 2013;8:e65522. , 
  • Poff AM, Ward N, Seyfried TN, et al. Non-toxic metabolic management of metastatic cancer in VM Mice: novel combination of ketogenic diet, ketone supplementation, and hyperbaric oxygen therapy. PLoS One. 2015;10:e0127407. , 
  • Ohguri T, Imada H, Narisada H, et al. Systemic chemotherapy using paclitaxel and carboplatin plus regional hyperthermia and hyperbaric oxygen treatment for non-small cell lung cancer with multiple pulmonary metastases: preliminary results. Int J Hyperthermia. 2009;25:160167. , 
  • Ohguri T, Kunugita N, Yahara K, et al. Efficacy of hyperbaric oxygen therapy combined with mild hyperthermia for improving the anti-tumour effects of carboplatin. Int J Hyperthermia. 2015;31:643648. , 
  • Xu MJ, Alberts DS. Potentiation of platinum analogue cytotoxicity by hyperthermia. Cancer Chemother Pharmacol. 1988;21:191196. , 
  • Herman TS, Teicher BA, Chan V, et al. Effect of heat on the cytotoxicity and interaction with DNA of a series of platinum complexes. Int J Radiat Oncol Biol Phys. 1989;16:443449. , 
  • Othman T, Goto S, Lee JB, et al. Hyperthermic enhancement of the apoptotic and antiproliferative activities of paclitaxel. Pharmacology. 2001;62:208212. , 
  • Cividalli A, Cruciani G, Livdi E, et al. Hyperthermia enhances the response of paclitaxel and radiation in a mouse adenocarcinoma. Int J Radiat Oncol Biol Phys. 1999;44:407412. , 
  • Zoul Z, Filip S, Melichar B, et al. Weekly paclitaxel combined with local hyperthermia in the therapy of breast cancer locally recurrent after mastectomy–a pilot experience. Oncol Res Treat. 2004;27:385388. , 
  • Moyer HR, Delman KA. The role of hyperthermia in optimizing tumor response to regional therapy. Int J Hyperthermia. 2008;24:251261. , 
  • Jones EL, Samulski TV, Vujaskovic Z, et al. Hyperthermia. In: Perez CA, Brady LW, Halperin WC, et al. editors. Principles and practice of radiation oncology. 4th ed. Philadelphia: Lippincott Williams & Wilkins; 2003. p. 699735.  
  • Wouters BG, van den BT, Magagnin MG, et al. Targeting hypoxia tolerance in cancer. Drug Resist Updat. 2004;7:2540. , 
  • Vaupel P, Thews O, Hoeckel M. Treatment resistance of solid tumors: role of hypoxia and anemia. MO. 2001;18:243259. , 
  • Hoogsteen IJ, Marres HA, van der Kogel AJ, et al. The hypoxic tumour microenvironment, patient selection and hypoxia-modifying treatments. Clin Oncol (R Coll Radiol). 2007;19:385396. , 
  • Vaupel P, Mayer A, Hockel M. Tumor hypoxia and malignant progression. Meth Enzymol. 2004;381:335354. , 
  • Vaupel P, Harrison L. Tumor hypoxia: causative factors, compensatory mechanisms, and cellular response. Oncologist. 2004;9:49. , 
  • Stuhr LE, Raa A, Oyan AM, et al. Hyperoxia retards growth and induces apoptosis, changes in vascular density and gene expression in transplanted gliomas in nude rats. J Neurooncol. 2007;85:191202. , 
  • Moen I, Oyan AM, Kalland KH, et al. Hyperoxic treatment induces mesenchymal-to-epithelial transition in a rat adenocarcinoma model. PLoS One. 2009;4:e6381. , 
  • Stuhr LE, Iversen VV, Straume O, et al. Hyperbaric oxygen alone or combined with 5-FU attenuates growth of DMBA-induced rat mammary tumors. Cancer Lett. 2004;210:3540. , 
  • Petre PM, Baciewicz FA, Jr., Tigan S, et al. Hyperbaric oxygen as a chemotherapy adjuvant in the treatment of metastatic lung tumors in a rat model. J Thorac Cardiovasc Surg. 2003;125:8595; discussion 95. , 
  • Al-Waili NS, Butler GJ, Beale J, et al. Hyperbaric oxygen and malignancies: a potential role in radiotherapy, chemotherapy, tumor surgery and phototherapy. Med Sci Monit. 2005;11:RA279RA289. , 
  • Bennett M, Feldmeier J, Smee R, et al. Hyperbaric oxygenation for tumour sensitisation to radiotherapy: a systematic review of randomised controlled trials. Cancer Treat Rev. 2008;34:577591. , 
  • Schwartz LH, Litiere S, de Vries E, et al. RECIST 1.1-Update and clarification: from the RECIST committee. Eur J Cancer. 2016;62:132137. , 
  • Common Terminology Criteria for Adverse Events (CTCAE): National Cancer Institute; 2010 [cited 2018 July 23]. Available from: https://ctep.cancer.gov/protocoldevelopment/electronic_applications/ctc.htm  
  • Belani CP, Barstis J, Perry MC, et al. Multicenter, randomized trial for stage IIIB or IV non-small-cell lung cancer using weekly paclitaxel and carboplatin followed by maintenance weekly paclitaxel or observation. JCO. 2003;21:29332939. , 
  • Belani CP, Ramalingam S, Perry MC, et al. Randomized, phase III study of weekly paclitaxel in combination with carboplatin versus standard every-3-weeks administration of carboplatin and paclitaxel for patients with previously untreated advanced non-small-cell lung cancer. JCO. 2008;26:468473. , 
  • Schuette W, Blankenburg T, Guschall W, et al. Multicenter randomized trial for stage IIIB/IV non-small-cell lung cancer using every-3-week versus weekly paclitaxel/carboplatin. Clin Lung Cancer. 2006;7:338343. , 
  • Liu SV, Camidge DR, Gettinger SN, et al. Long-term survival follow-up of atezolizumab in combination with platinum-based doublet chemotherapy in patients with advanced non-small-cell lung cancer. Eur J Cancer. 2018;101:114122. , 
  • Gillies RJ, Robey I, Gatenby RA. Causes and consequences of increased glucose metabolism of cancers. J Nucl Med. 2008;49: 24S42S. , 
  • Frezza C, Pollard PJ, Gottlieb E. Inborn and acquired metabolic defects in cancer. J Mol Med. 2011;89:213220. , 
  • Bayley JP, Devilee P. The Warburg effect in 2012. Curr Opin Oncol. 2012;24:6267. , 
  • Shinitzky M, Henkart P. Fluidity of cell membranes-current concepts and trends. Int Rev Cytol. 1979;60:121147. , 
  • Demetrius LA, Coy JF, Tuszynski JA. Cancer proliferation and therapy: the Warburg effect and quantum metabolism. Theor Biol Med Model. 2010;7:2. , 
  • Schilsky RL, Bailey BD, Chabner BA. Characteristics of membrane transport of methotrexate by cultured human breast cancer cells. Biochem Pharmacol. 1981;30:15371542. , 
  • Gasparro FP, Krobler RM, Yemul SS, et al. Receptor-mediated photo-cytotoxicity: synthesis of a photoactivatable psoralen derivative conjugated to insulin. Biochem Biophys Res Commun. 1986;141:502509. , 
  • Yoshimasa Y, Namba Y, Hanaoka M, et al. A new approach to the detection of autoantibodies against insulin receptors that inhibit the internalization of insulin into human cells. Diabetes. 1984;33:10511054. , 
  • Poznansky MJ, Singh R, Singh B, et al. Insulin: carrier potential for enzyme and drug therapy. Science. 1984;223:13041306. , 
  • Jeffcoat R. The biosynthesis of unsaturated fatty acids and its control in mammalian liver. Essays Biochem. 1979;15:136. , 
  • Papa V, Pezzino V, Costantino A, et al. Elevated insulin receptor content in human breast cancer. J Clin Invest. 1990;86:15031510. , 
  • Yee D. The insulin-like growth factors and breast cancer-revisited. Breast Cancer Res Treat. 1998;47:197199. , 
  • Gross GE, Boldt DH, Osborne CK. Perturbation by insulin of human breast cancer cell cycle kinetics. Cancer Res. 1984;44:35703575. , 
  • Toth C, Clemens Z. Halted progression of soft palate cancer in a patient treated with the paleolithic ketogenic diet alone: a 20-months follow-up. Am J Med Case Rep. 2016;4:288292.  
  • Schmidt M, Pfetzer N, Schwab M, et al. Effects of a ketogenic diet on the quality of life in 16 patients with advanced cancer: a pilot trial. Nutr Metab (Lond). 2011;8:54. , 
  • Rieger J, Bahr O, Maurer GD, et al. ERGO: a pilot study of ketogenic diet in recurrent glioblastoma. Int J Oncol. 2014;44:18431852. , 
  • Seyfried TN. Case studies and personal experiences in using the ketogenic diet for cancer management, in cancer as a metabolic disease: on the origin, management and prevention of cancer. Hoboken, NJ, USA: John Wiley & Sons, Inc.; 2012. , 
  • Fine EJ, Segal-Isaacson CJ, Feinman RD, et al. Targeting insulin inhibition as a metabolic therapy in advanced cancer: a pilot safety and feasibility dietary trial in 10 patients. Nutrition. 2012;28:10281035. , 
  • Champ CE, Palmer JD, Volek JS, et al. Targeting metabolism with a ketogenic diet during the treatment of glioblastoma multiforme. J Neurooncol. 2014;117:125131. , 
  • Martinez-Outschoorn UE, Lin Z, Whitaker-Menezes D, et al. Ketone body utilization drives tumor growth and metastasis. Cell Cycle. 2012;11:39643971. , 
  • Martinez-Outschoorn UE, Prisco M, Ertel A, et al. Ketones and lactate increase cancer cell "stemness," driving recurrence, metastasis and poor clinical outcome in breast cancer: achieving personalized medicine via Metabolo-Genomics. Cell Cycle. 2011;10:12711286. , 
  • Bonuccelli G, Tsirigos A, Whitaker-Menezes D, et al. Ketones and lactate "fuel" tumor growth and metastasis: evidence that epithelial cancer cells use oxidative mitochondrial metabolism. Cell Cycle. 2010;9:35063514. , 
  • Tisdale MJ, Brennan RA. Loss of acetoacetate coenzyme A transferase activity in tumours of peripheral tissues. Br J Cancer. 1983;47:293297. , 
  • Sawai M, Yashiro M, Nishiguchi Y, et al. Growth-inhibitory effects of the ketone body, monoacetoacetin, on human gastric cancer cells with succinyl-CoA: 3-oxoacid CoA-transferase (SCOT) deficiency. Anticancer Res. 2004;24:22132217. , 
  • Maurer GD, Brucker DP, Bahr O, et al. Differential utilization of ketone bodies by neurons and glioma cell lines: a rationale for ketogenic diet as experimental glioma therapy. BMC Cancer. 2011;11:315. , 
  • Skinner R, Trujillo A, Ma X, et al. Ketone bodies inhibit the viability of human neuroblastoma cells. J Pediatr Surg. 2009;44:212216. discussion 216. , 
  • Klement RJ, Champ CE, Otto C, et al. Anti-tumor effects of ketogenic diets in mice: a meta-analysis. PLoS One. 2016;11:e0155050. , 
  • Magee BA, Potezny N, Rofe AM, et al. The inhibition of malignant cell growth by ketone bodies. Aust J Exp Biol Med Sci. 1979;57:529539. , 
  • Poff AM, Ari C, Arnold P, et al. Ketone supplementation decreases tumor cell viability and prolongs survival of mice with metastatic cancer. Int J Cancer. 2014;135:17111720. , 
  • Abdelwahab MG, Fenton KE, Preul MC, et al. The ketogenic diet is an effective adjuvant to radiation therapy for the treatment of malignant glioma. PLoS One. 2012;7:e36197. , 
  • Schwartz K, Chang HT, Nikolai M, et al. Treatment of glioma patients with ketogenic diets: report of two cases treated with an IRB-approved energy-restricted ketogenic diet protocol and review of the literature. Cancer Metab. 2015;3:3. , 
  • Woolf EC, Scheck AC. The ketogenic diet for the treatment of malignant glioma. J Lipid Res. 2015;56:510. , 
  • Seyfried TN, Yu G, Maroon JC, et al. Press-pulse: a novel therapeutic strategy for the metabolic management of cancer. Nutr Metab (Lond). 2017;14:19. , 


Plaats een reactie ...

Reageer op "Hyperthermie plus voedingsprogramma (kytogeen dieet) plus hyperbare zuurstoftherapie aanvullend op chemo geeft uitstekende resultaten op mediane overall overleving (42 maanden) bij patienten met gevorderde niet-kleincellige longkanker"


Gerelateerde artikelen
 

Gerelateerde artikelen

Hyperthermie in combinatie >> Matters studie onderzoekt >> Electro hyperthermie naast >> Is de Hyperthermie infra rode >> Hyperthermie plus hoge dosis >> Hyperthermie heeft vaak therapeutisch >> Hyperthermie plus voedingsprogramma >> Hyperthermie studies in overzichtelijk >> Hyperthermie aanvullend op >> Algemeen: een vergelijking >>