Feasibility, Safety, and Preliminary Efficacy of Very Low-Volume Interval Training in Advanced Cancer Patients

Dejan Reljic, Hans J Herrmann, Benedikt Jakobs, Walburga Dieterich, Dimitrios Mougiakakos, Markus F Neurath, Yurdagül Zopf, Dejan Reljic, Hans J Herrmann, Benedikt Jakobs, Walburga Dieterich, Dimitrios Mougiakakos, Markus F Neurath, Yurdagül Zopf

Abstract

Purpose: High-intensity interval training (HIIT) has been shown to improve cardiorespiratory fitness (CRF) and health-related outcomes in various chronic diseases, including cancer. However, data on feasibility and efficacy of HIIT in advanced cancer patients are still sparse, presumably because of safety concerns, like suspected immunosuppression after vigorous exercise. This randomized, sham-intervention controlled study aimed to investigate feasibility, safety, and preliminary efficacy of very low-volume HIIT (LOW-HIIT) in advanced cancer patients.

Methods: Twenty-seven patients (55.4 ± 13.2 yr) with different advanced cancers (Union for International Cancer Control [UICC] III/IV) were randomly allocated to LOW-HIIT ( n = 13), consisting of 5 × 1 min cycle ergometer intervals (14 min per session total duration) at 80% to 95% HR peak (two sessions per week for 12 wk), or a sham intervention ( n = 14) performing light physical mobilization exercises (SHAM). Primary outcomes were attrition and attendance rates, with values of ≤25% and ≥80%, respectively, considered acceptable. Secondary outcomes were safety, protocol fidelity, physiological (including CRF measures) and patient-reported outcomes (including fatigue and quality of life).

Results: One of 13 patients (8%) receiving LOW-HIIT dropped out. Mean attendance rate was ~93%. The prescribed minimum exercise intensity was consistently reached by all patients. Low-volume HIIT was well tolerated and not associated with any serious adverse event nor with increased infection susceptibility. There were no biochemical signs of acute immunosuppression after LOW-HIIT. Contrarily, differentiation and degranulation of natural killer cells was acutely increased postexercise. Low-volume HIIT improved CRF measures including peak oxygen uptake, self-reported fatigue, physical, and social functioning. No significant changes occurred in the SHAM group.

Conclusions: Low-volume HIIT can be regarded as feasible and safe in advanced cancer patients. Our preliminary data indicate favorable acute effects on NK-cells and beneficial chronic adaptations in CRF, fatigue, and aspects of quality of life.

Trial registration: ClinicalTrials.gov NCT04065815.

Copyright © 2022 The Author(s). Published by Wolters Kluwer Health, Inc. on behalf of the American College of Sports Medicine.

Figures

FIGURE 1
FIGURE 1
Schematic illustration of study design. Blood draws in week 4 and week 8 before exercise session.
FIGURE 2
FIGURE 2
Study flowchart.
FIGURE 5
FIGURE 5
Individual heart rate responses and chronic adaptations to LOW-HIIT. Individual mean heart rate responses during exercise intervals in weeks 1 to 4, weeks 5 to 8, and weeks 9 to 12 (full line indicates minimum required heart rate [≥80% HRpeak), dashed lines indicate lower bounds of the intended heart rates for weeks 5 to 8 [≥85% HRpeak] and week 9 to 12 [≥90% HRpeak] (A), individual changes in maximal oxygen uptake (B), individual changes in VT (C), individual changes in FACIT score (D), individual changes in physical functioning (E), and individual changes in social functioning (F).
FIGURE 3
FIGURE 3
Acute responses of leukocytes (A), lymphocytes (B), monocytes (C), and neutrophils (D) to LOW-HIIT.
FIGURE 4
FIGURE 4
Acute responses of NK cells to LOW-HIIT. NK cell degranulation after contact with K562 and BL-41 cells within 60-min postexercise (A), NK cell degranulation preexercise vs 60-min postexercise (B), NK cell differentiation within 60-min postexercise (C), and NK cell differentiation preexercise vs directly postexercise (D). *Significant difference (P < 0.05) preexercise vs. 60-min postexercise; **Significant difference (P < 0.01) preexercise vs directly postexercise.

References

    1. Moore SC Lee IM Weiderpass E, et al. . Association of leisure-time physical activity with risk of 26 types of cancer in 1.44 million adults. JAMA Intern Med. 2016;176(6):816–25.
    1. Campbell KL Winters-Stone KM Wiskemann J, et al. . Exercise guidelines for cancer survivors: consensus statement from international multidisciplinary roundtable. Med Sci Sports Exerc. 2019;51(11):2375–90.
    1. Heywood R, McCarthy AL, Skinner TL. Safety and feasibility of exercise interventions in patients with advanced cancer: a systematic review. Support Care Cancer. 2017;25(10):3031–50.
    1. Weller S Hart NH Bolam KA, et al. . Exercise for individuals with bone metastases: a systematic review. Crit Rev Oncol Hematol. 2021;166:103433.
    1. Sheill G, Guinan E, Brady L, Hevey D, Hussey J. Exercise interventions for patients with advanced cancer: a systematic review of recruitment, attrition, and exercise adherence rates. Palliat Support Care. 2019;17(6):686–96.
    1. Albrecht TA, Taylor AG. Physical activity in patients with advanced-stage cancer: a systematic review of the literature. Clin J Oncol Nurs. 2012;16(3):293–300.
    1. Gilliam LAA, St Clair DK. Chemotherapy-induced weakness and fatigue in skeletal muscle: the role of oxidative stress. Antioxid Redox Signal. 2011;15(9):2543–63.
    1. Foulkes SJ Howden EJ Bigaran A, et al. . Persistent impairment in cardiopulmonary fitness after breast cancer chemotherapy. Med Sci Sports Exerc. 2019;51(8):1573–81.
    1. Murphy CL, Sheane BJ, Cunnane G. Attitudes towards exercise in patients with chronic disease: the influence of comorbid factors on motivation and ability to exercise. Postgrad Med J. 2011;87(1024):96–100.
    1. Gibala MJ, Gillen JB, Percival ME. Physiological and health-related adaptations to low-volume interval training: influences of nutrition and sex. Sports Med. 2014;44(2 Suppl):127–37.
    1. Mattioni Maturana F, Martus P, Zipfel S, Nieß AM. Effectiveness of HIIE versus MICT in improving cardiometabolic risk factors in health and disease: a meta-analysis. Med Sci Sports Exerc. 2021;53(3):559–73.
    1. Mugele H Freitag N Wilhelmi J, et al. . High-intensity interval training in the therapy and aftercare of cancer patients: a systematic review with meta-analysis. J Cancer Surviv. 2019;13(2):205–23.
    1. Palma S, Hasenoehrl T, Jordakieva G, Ramazanova D, Crevenna R. High-intensity interval training in the prehabilitation of cancer patients-a systematic review and meta-analysis. Support Care Cancer. 2021;29(4):1781–94.
    1. Tsuji K, Matsuoka YJ, Ochi E. High-intensity interval training in breast cancer survivors: a systematic review. BMC Cancer. 2021;21(1):184.
    1. Wallen MP Hennessy D Brown S, et al. . High-intensity interval training improves cardiorespiratory fitness in cancer patients and survivors: a meta-analysis. Eur J Cancer Care. 2020;29(4):e13267.
    1. Souza D Vale AF Silva A, et al. . Acute and chronic effects of interval training on the immune system: a systematic review with meta-analysis. Biology (Basel). 2021;10(9):868.
    1. Llavero F Alejo LB Fiuza-Luces C, et al. . Exercise training effects on natural killer cells: a preliminary proteomics and systems biology approach. Exerc Immunol Rev. 2021;27:125–41.
    1. Campbell JP, Turner JE. There is limited existing evidence to support the common assumption that strenuous endurance exercise bouts impair immune competency. Expert Rev Clin Immunol. 2019;15(2):105–9.
    1. Rabinovich GA, Gabrilovich D, Sotomayor EM. Immunosuppressive strategies that are mediated by tumor cells. Annu Rev Immunol. 2007;25:267–96.
    1. Reljic D, Wittmann F, Fischer JE. Effects of low-volume high-intensity interval training in a community setting: a pilot study. Eur J Appl Physiol. 2018;118(6):1153–67.
    1. Reljic D, Frenk F, Herrmann HJ, Neurath MF, Zopf Y. Effects of very low volume high intensity versus moderate intensity interval training in obese metabolic syndrome patients: a randomized controlled study. Sci Rep. 2021;11(1):2836.
    1. Reljic D, Frenk F, Herrmann HJ, Neurath MF, Zopf Y. Low-volume high-intensity interval training improves cardiometabolic health, work ability and well-being in severely obese individuals: a randomized-controlled trial sub-study. J Transl Med. 2020;18(1):419.
    1. Julious SA. Sample size of 12 per group rule of thumb for a pilot study. Pharm Stat. 2005;4(4):287–91.
    1. National Cancer Institute Web site [Internet] . Rockville (MD): National Cancer Institute; [cited 2022 April 6]. Available from: .
    1. Vicent L Ariza-Solé A González-Juanatey JR, et al. . Exercise-related severe cardiac events. Scand J Med Sci Sports. 2018;28(4):1404–11.
    1. National Cancer Institute Web site [Internet] . Rockville (MD): National Cancer Institute; [cited 2022 April 6]. Available from: .
    1. Matomäki P, Kainulainen H, Kyröläinen H. Corrected whole blood biomarkers - the equation of dill and Costill revisited. Phys Rep. 2018;6(12):e13749.
    1. Bosy-Westphal A, Jensen B, Braun W, Pourhassan M, Gallagher D, Müller MJ. Quantification of whole-body and segmental skeletal muscle mass using phase-sensitive 8-electrode medical bioelectrical impedance devices. Eur J Clin Nutr. 2017;71(9):1061–7.
    1. Howley ET, Bassett DR, Jr, Welch HG. Criteria for maximal oxygen uptake: review and commentary. Med Sci Sports Exerc. 1995;27(9):1292–301.
    1. Meyer T, Lucía A, Earnest CP, Kindermann W. A conceptual framework for performance diagnosis and training prescription from submaximal gas exchange parameters—theory and application. Int J Sports Med. 2005;26(1 Suppl):S38–48.
    1. Montan I, Löwe B, Cella D, Mehnert A, Hinz A. General population norms for the functional assessment of chronic illness therapy (FACIT)—fatigue scale. Value Health. 2018;21(11):1313–21.
    1. Singer S Wollbrück D Wulke C, et al. . Validation of the EORTC QLQ-C30 and EORTC QLQ-H&N35 in patients with laryngeal cancer after surgery. Head Neck. 2009;31(1):64–76.
    1. Karnofsky DA, Burchenal JH. “The Clinical Evaluation of Chemotherapeutic Agents in Cancer,” in Evaluation of Chemotherapeutic Agents. New York, NY: Columbia University Press; 1949. pp. 191–205.
    1. Arends J Bachmann P Baracos V, et al. . ESPEN guidelines on nutrition in cancer patients. Clin Nutr. 2017;36(1):11–48.
    1. Schofield WN. Predicting basal metabolic rate, new standards and review of previous work. Hum Nutr Clin Nutr. 1985;39(1 Suppl):5–41.
    1. Cohen J. Statistical Power Analysis for the Behavioral Sciences. 2nd ed. Routledge, UK: Taylor and Francis; 1988.
    1. Reljic D, Lampe D, Wolf F, Zopf Y, Herrmann HJ, Fischer J. Prevalence and predictors of dropout from high-intensity interval training in sedentary individuals: a meta-analysis. Scand J Med Sci Sports. 2019;29(9):1288–304.
    1. Knox ECL, Webb OJ, Esliger DW, Biddle SJH, Sherar LB. Using threshold messages to promote physical activity: implications for public perceptions of health effects. Eur J Pub Health. 2014;24(2):195–9.
    1. Quist M Rørth M Langer S, et al. . Safety and feasibility of a combined exercise intervention for inoperable lung cancer patients undergoing chemotherapy: a pilot study. Lung Cancer. 2012;75(2):203–8.
    1. Van den Dungen IA, Verhagen CA, van der Graaf WT, van den Berg JP, Vissers KC, Engels Y. Feasibility and impact of a physical exercise program in patients with advanced cancer: a pilot study. J Palliat Med. 2014;17(10):1091–8.
    1. Cooper MA, Fehniger TA, Caligiuri MA. The biology of human natural killer-cell subsets. Trends Immunol. 2001;22(11):633–40.
    1. Idorn M, Hojman P. Exercise-dependent regulation of NK cells in cancer protection. Trends Mol Med. 2016;22(7):565–77.
    1. Bigley AB Rezvani K Chew C, et al. . Acute exercise preferentially redeploys NK-cells with a highly-differentiated phenotype and augments cytotoxicity against lymphoma and multiple myeloma target cells. Brain Behav Immun. 2014;39:160–71.
    1. Barra NG Fan IY Gillen JB, et al. . High intensity interval training increases natural killer cell number and function in obese breast cancer-challenged mice and obese women. J Cancer Prev. 2017;22(4):260–6.
    1. Fardman A Banschick GD Rabia R, et al. . Cardiorespiratory fitness and survival following cancer diagnosis. Eur J Prev Cardiol. 2021;28(11):1242–9.
    1. Imboden MT Harber MP Whaley MH, et al. . The association between the change in directly measured cardiorespiratory fitness across time and mortality risk. Prog Cardiovasc Dis. 2019;62(2):157–62.
    1. Sinclair RCF, Sumpter K, Griffin SM. Fitness after chemotherapy. Br J Anaesth. 2016;116(1):140.
    1. Nakamura T, Matsumine A, Matsubara T, Asanuma K, Uchida A, Sudo A. Clinical significance of pretreatment serum C-reactive protein level in soft tissue sarcoma. Cancer. 2012;118(4):1055–61.
    1. Gillison FB, Skevington SM, Sato A, Standage M, Evangelidou S. The effects of exercise interventions on quality of life in clinical and healthy populations; a meta-analysis. Soc Sci Med. 2009;68(9):1700–10.
    1. Cella D Wilson H Shalhoub H, et al. . Content validity and psychometric evaluation of functional assessment of chronic illness therapy—fatigue in patients with psoriatic arthritis. J Patient Rep Outcomes. 2019;3(1):30.

Source: PubMed

3
Suscribir