Nutritional Counseling During Chemotherapy Treatment: A Systematic Review of Feasibility, Safety, and Efficacy
Abstract
:1. Introduction
2. Methods
2.1. Eligibility Criteria
2.2. Search Strategy
2.3. Data Extraction and Risk of Bias Assessment
3. Results
3.1. Study Design
3.2. Study Characteristics
3.3. Nutritional Interventions
3.4. Feasibility
3.5. Safety
3.6. Primary Health Outcomes Assessed
3.7. Efficacy
3.7.1. Body Weight or Body Composition
3.7.2. Nutritional Status
3.7.3. Quality of Life
3.7.4. Cancer-Related Fatigue
3.7.5. Chemotherapy Tolerance
3.7.6. Response to Chemotherapy
3.7.7. Other Outcomes
4. Discussion
5. Conclusions
Supplementary Materials
Author Contributions
Funding
Institutional Review Board Statement
Data Availability Statement
Conflicts of Interest
References
- World Health Organization; International Agency for Research on Cancer. Cancer Fact Sheets, All Cancers; Globocan: Lyon, France, 2022. [Google Scholar]
- Nekhlyudov, L.; Ganz, P.A.; Arora, N.K.; Rowland, J.H. Going beyong being lost in transition: A decade of progress in cancer survivorship. J. Clin. Oncol. 2017, 35, 1978–1981. [Google Scholar] [CrossRef]
- Arends, J.; Bachmann, P.; Baracos, V.; Barthelemy, N.; Bertz, H.; Bozzetti, F.; Fearon, K.; Hütterer, E.; Isenring, E.; Kaasa, S.; et al. ESPEN guidelines on nutrition in cancer patients. Clin. Nutr. 2017, 36, 11–48. [Google Scholar] [CrossRef] [PubMed]
- de van der Schueren, M.A.E.; Laviano, A.; Blanchard, H.; Jourdan, M.; Arends, J.; Baracos, V.E. Systematic review and meta-analysis of the evidence for oral nutritional intervention on nutritional and clinical outcomes during chemo(radio)therapy: Current evidence and guidance for design of future trials. Ann. Oncol. 2018, 29, 1141–1153. [Google Scholar] [CrossRef]
- Brown, J.K.; Byers, T.; Doyle, C.; Coumeya, K.S.; Demark-Wahnefried, W.; Kushi, L.H.; McTieman, A.; Rock, C.L.; Aziz, N.; Bloch, A.S.; et al. Nutrition and physical activity during and after cancer treatment: An American Cancer Society guide for informed choices. CA Cancer J. Clin. 2003, 53, 268–291. [Google Scholar] [CrossRef]
- Harvey, B.I.; Youngblood, S.M.; Kleckner, A.S. Barriers and Facilitators to Adherence to a Mediterranean Diet Intervention during Chemotherapy Treatment: A Qualitative Descriptive Analysis. Nutr. Cancer 2023, 75, 1349–1360. [Google Scholar] [CrossRef]
- Rock, C.L.; Thomson, C.A.; Sullivan, K.R.; Howe, C.L.; Kushi, L.H.; Caan, B.J.; Neuhouser, M.L.; Bandera, E.V.; Wang, Y.; Robien, K.; et al. American Cancer Society nutrition and physical activity guideline for cancer survivors. CA Cancer J. Clin. 2022, 72, 230–262. [Google Scholar] [CrossRef]
- Ligibel, J.A.; Bohlke, K.; May, A.M.; Clinton, S.K.; Demark-Wahnefried, W.; Gilchrist, S.C.; Irwin, M.L.; Late, M.; Mansfield, S.; Marshall, T.F.; et al. Exercise, Diet, and Weight Management During Cancer Treatment: ASCO Guideline. J. Clin. Oncol. 2022, 40, 2491–2507. [Google Scholar] [CrossRef]
- Arends, J.; Baracos, V.; Bertz, H.; Bozzetti, F.; Calder, P.C.; Deutz, N.E.P.; Erickson, N.; Laviano, A.; Lisanti, M.P.; Lobo, D.N.; et al. ESPEN expert group recommendations for action against cancer-related malnutrition. Clin. Nutr. 2017, 36, 1187–1196. [Google Scholar] [CrossRef]
- Ambrosone, C.B.; Zirpoli, G.R.; Hutson, A.D.; McCann, W.E.; McCann, S.E.; Barlow, W.E.; Kelly, K.M.; Cannioto, R.; Sucheston-Campbell, L.E.; Hershman, D.L.; et al. Dietary supplement use during chemotherapy and survival outcomes of patients with breast cancer enrolled in a cooperative group clinical trial (SWOG S0221). J. Clin. Oncol. 2020, 38, 804–814. [Google Scholar] [CrossRef]
- Kirkham, A.A.; Parr, E.B.; Kleckner, A.S. Cardiometabolic health impacts of time-restricted eating: Implications for type 2 diabetes, cancer, and cardiovascular diseases. Curr. Opin. Clin. Nutr. Metab. Care 2022, 25, 378–387. [Google Scholar] [CrossRef] [PubMed]
- Kalam, F.; James, D.; Li, Y.R.; Coleman, M.F.; Kiesel, V.A.; Cespedes Feliciano, E.M.; Hursting, S.D.; Sears, D.D.; Kleckner, A.S. Intermittent fasting interventions to leverage chrononutrition and metabolic mechanisms for cancer treatment and supportive care outcomes. JNCI Monogr. 2023, 61, 84–103. [Google Scholar] [CrossRef] [PubMed]
- Choi, J.; Zhang, X.; Li, W.; Houston, M.; Peregrina, K.; Dubin, R.; Ye, K.; Augenlicht, L. Dynamic Intestinal Stem Cell Plasticity and Lineage Remodeling by a Nutritional Environment Relevant to Human Risk for Tumorigenesis. Mol. Cancer Res. 2023, 21, 808–824. [Google Scholar] [CrossRef] [PubMed]
- Li, W.; Zimmerman, S.E.; Peregrina, K.; Houston, M.; Mayoral, J.; Zhang, J.; Maqbool, S.; Zhang, Z.; Cai, Y.; Ye, K.; et al. The nutritional environment determines which and how intestinal stem cells contribute to homeostasis and tumorigenesis. Carcinogenesis 2019, 40, 937–946. [Google Scholar] [CrossRef]
- Yee, L.D.; Lester, J.L.; Cole, R.M.; Richardson, J.R.; Hsu, J.C.; Li, Y.; Lehman, A.; Belury, M.A.; Clinton, S.K. Omega-3 fatty acid supplements in women at high risk of breast cancer have dose-dependent effects on breast adipose tissue fatty acid composition. Am. J. Clin. Nutr. 2010, 91, 1185–1194. [Google Scholar] [CrossRef]
- Straka, S.; Lester, J.L.; Cole, R.M.; Andridge, R.R.; Puchala, S.; Rose, A.M.; Clinton, S.K.; Belury, M.A.; Yee, L.D. Incorporation of eicosapentaenioic and docosahexaenoic acids into breast adipose tissue of women at high risk of breast cancer: A randomized clinical trial of dietary fish and n-3 fatty acid capsules. Mol. Nutr. Food Res. 2015, 59, 1780–1790. [Google Scholar] [CrossRef] [PubMed]
- Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E.; et al. The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. BMJ 2021, 372, n71. [Google Scholar] [CrossRef] [PubMed]
- Sterne, J.A.C.; Savovic, J.; Page, M.J.; Elbers, R.G.; Blencowe, N.S.; Boutron, I.; Cates, C.J.; Cheng, H.Y.; Corbett, M.S.; Eldridge, S.M.; et al. RoB 2: A revised tool for assessing risk of bias in randomised trials. BMJ 2019, 366, l4898. [Google Scholar] [CrossRef] [PubMed]
- Abdollahi, R.; Najafi, S.; Razmpoosh, E.; Shoormasti, R.S.; Haghighat, S.; Raji Lahiji, M.; Chamari, M.; Asgari, M.; Cheshmazar, E.; Zarrati, M. The Effect of Dietary Intervention Along with Nutritional Education on Reducing the Gastrointestinal Side Effects Caused by Chemotherapy Among Women with Breast Cancer. Nutr. Cancer 2019, 71, 922–930. [Google Scholar] [CrossRef] [PubMed]
- Baldwin, C.; Spiro, A.; McGough, C.; Norman, A.R.; Gillbanks, A.; Thomas, K.; Cunningham, D.; O’Brien, M.; Andreyev, H.J. Simple nutritional intervention in patients with advanced cancers of the gastrointestinal tract, non-small cell lung cancers or mesothelioma and weight loss receiving chemotherapy: A randomised controlled trial. J. Hum. Nutr. Diet. 2011, 24, 431–440. [Google Scholar] [CrossRef]
- Bourdel-Marchasson, I.; Blanc-Bisson, C.; Doussau, A.; Germain, C.; Blanc, J.F.; Dauba, J.; Lahmar, C.; Terrebonne, E.; Lecaille, C.; Ceccaldi, J.; et al. Nutritional advice in older patients at risk of malnutrition during treatment for chemotherapy: A two-year randomized controlled trial. PLoS ONE 2014, 9, e108687. [Google Scholar] [CrossRef]
- Dai, W.; Wang, S.A.; Wang, K.; Chen, C.; Wang, J.; Chen, X.; Yan, J. Impact of Nutrition Counseling in Head and Neck Cancer Sufferers Undergoing Antineoplastic Therapy: A Randomized Controlled Pilot Study. Curr. Oncol. 2022, 29, 6947–6955. [Google Scholar] [CrossRef]
- de Lima Bezerra, A.D.; Matias de Sousa, I.; Silva de Souza, A.P.; Miranda de Carvalho, A.L.; Trussardi Fayh, A.P. Early nutritional intervention does not prevent long-term adverse events in women with breast cancer: A pilot study. Clin. Nutr. ESPEN 2023, 53, 268–273. [Google Scholar] [CrossRef] [PubMed]
- de Souza, A.P.S.; Silva, L.C.D.; Fayh, A.P.T. Nutritional Intervention Contributes to the Improvement of Symptoms Related to Quality of Life in Breast Cancer Patients Undergoing Neoadjuvant Chemotherapy: A Randomized Clinical Trial. Nutrients 2021, 13, 589. [Google Scholar] [CrossRef]
- Keum, J.; Chung, M.J.; Kim, Y.; Ko, H.; Sung, M.J.; Jo, J.H.; Park, J.Y.; Bang, S.; Park, S.W.; Song, S.Y.; et al. Usefulness of Smartphone Apps for Improving Nutritional Status of Pancreatic Cancer Patients: Randomized Controlled Trial. JMIR Mhealth Uhealth 2021, 9, e21088. [Google Scholar] [CrossRef]
- Lin, J.X.; Chen, X.W.; Chen, Z.H.; Huang, X.Y.; Yang, J.J.; Xing, Y.F.; Yin, L.H.; Li, X.; Wu, X.Y. A multidisciplinary team approach for nutritional interventions conducted by specialist nurses in patients with advanced colorectal cancer undergoing chemotherapy: A clinical trial. Medicine 2017, 96, e7373. [Google Scholar] [CrossRef] [PubMed]
- Loprinzi, C.L.; Athmann, L.M.; Kardinal, C.G.; O’Fallon, J.R.; See, J.A.; Bruce, B.K.; Dose, A.M.; Miser, A.W.; Kern, P.S.; Tschetter, L.K.; et al. Randomized trial of dietician counseling to try to prevent weight gain associated with breast cancer adjuvant chemotherapy. Oncology 1996, 53, 228–232. [Google Scholar] [CrossRef] [PubMed]
- Najafi, S.; Haghighat, S.; Raji Lahiji, M.; RazmPoosh, E.; Chamari, M.; Abdollahi, R.; Asgari, M.; Zarrati, M. Randomized Study of the Effect of Dietary Counseling During Adjuvant Chemotherapy on Chemotherapy Induced Nausea and Vomiting, and Quality of Life in Patients With Breast Cancer. Nutr. Cancer 2019, 71, 575–584. [Google Scholar] [CrossRef] [PubMed]
- Ollenschlager, G.; Thomas, W.; Konkol, K.; Diehl, V.; Roth, E. Nutritional behaviour and quality of life during oncological polychemotherapy: Results of a prospective study on the efficacy of oral nutrition therapy in patients with acute leukaemia. Eur. J. Clin. Investig. 1992, 22, 546–553. [Google Scholar] [CrossRef]
- Ovesen, L.; Allingstrup, L.; Hannibal, J.; Mortensen, E.L.; Hansen, O.P. Effects of dietary counseling on food intake, body weight, response rate, survival, and quality of life in cancer patients undergoing chemotherapy: A prospective, randomized study. J. Clin. Oncol. 1993, 11, 2043–2049. [Google Scholar] [CrossRef]
- Regueme, S.C.; Echeverria, I.; Moneger, N.; Durrieu, J.; Becerro-Hallard, M.; Duc, S.; Lafargue, A.; Mertens, C.; Laksir, H.; Ceccaldi, J.; et al. Protein intake, weight loss, dietary intervention, and worsening of quality of life in older patients during chemotherapy for cancer. Support. Care Cancer 2021, 29, 687–696. [Google Scholar] [CrossRef]
- Sukaraphat, N.; Chewaskulyong, B.; Buranapin, S. Dietary counseling outcomes in locally advanced unresectable or metastatic cancer patients undergoing chemotherapy. J. Med. Assoc. Thail. 2016, 99, 1283–1290. [Google Scholar]
- van der Werf, A.; Langius, J.A.E.; Beeker, A.; Ten Tije, A.J.; Vulink, A.J.; Haringhuizen, A.; Berkhof, J.; van der Vliet, H.J.; Verheul, H.M.W.; de van der Schueren, M.A.E. The effect of nutritional counseling on muscle mass and treatment outcome in patients with metastatic colorectal cancer undergoing chemotherapy: A randomized controlled trial. Clin. Nutr. 2020, 39, 3005–3013. [Google Scholar] [CrossRef] [PubMed]
- Wang, S.A.; Li, F.; Zhu, J.; Chen, X.; Ren, W.; Gao, B. Multidisciplinary nutritional management improves nutritional and hospitalized outcomes of patients with esophageal cancer undergoing chemoradiotherapy: A randomized control trial. Medicine 2023, 102, e33335. [Google Scholar] [CrossRef]
- White, K.L.; Henson, C.C.; Hann, M.; Eden, M.; Burden, S.T.; Lal, S.; Davidson, S.E.; McLaughlin, J.T. Randomised clinical trial of a gastrointestinal care bundle to reduce symptoms in patients with pelvic cancer undergoing chemoradiotherapy. BMJ Open Gastroenterol. 2020, 7, e000432. [Google Scholar] [CrossRef] [PubMed]
- Xie, F.L.; Wang, Y.Q.; Peng, L.F.; Lin, F.Y.; He, Y.L.; Jiang, Z.Q. Beneficial Effect of Educational and Nutritional Intervention on the Nutritional Status and Compliance of Gastric Cancer Patients Undergoing Chemotherapy: A Randomized Trial. Nutr. Cancer 2017, 69, 762–771. [Google Scholar] [CrossRef] [PubMed]
- Allen, S.K.; Brown, V.; White, D.; King, D.; Hunt, J.; Wainwright, J.; Emery, A.; Hodge, E.; Kehinde, A.; Prabhu, P.; et al. Multimodal Prehabilitation During Neoadjuvant Therapy Prior to Esophagogastric Cancer Resection: Effect on Cardiopulmonary Exercise Test Performance, Muscle Mass and Quality of Life—A Pilot Randomized Clinical Trial. Ann. Surg. Oncol. 2022, 29, 1839–1850. [Google Scholar] [CrossRef]
- Basen-Engquist, K.M.; Raber, M.; Carmack, C.L.; Arun, B.; Brewster, A.M.; Fingeret, M.; Schembre, S.M.; Harrison, C.; Perkins, H.Y.; Li, Y.; et al. Feasibility and efficacy of a weight gain prevention intervention for breast cancer patients receiving neoadjuvant chemotherapy: A randomized controlled pilot study. Support. Care Cancer 2020, 28, 5821–5832. [Google Scholar] [CrossRef]
- Carayol, M.; Ninot, G.; Senesse, P.; Bleuse, J.P.; Gourgou, S.; Sancho-Garnier, H.; Sari, C.; Romieu, I.; Romieu, G.; Jacot, W. Short- and long-term impact of adapted physical activity and diet counseling during adjuvant breast cancer therapy: The “APAD1” randomized controlled trial. BMC Cancer 2019, 19, 737. [Google Scholar] [CrossRef] [PubMed]
- Demark-Wahnefried, W.; Case, L.D.; Blackwell, K.; Marcom, P.K.; Kraus, W.; Aziz, N.; Snyder, D.C.; Giguere, J.K.; Shaw, E. Results of a diet/exercise feasibility trial to prevent adverse body composition change in breast cancer patients on adjuvant chemotherapy. Clin. Breast Cancer 2008, 8, 70–79. [Google Scholar] [CrossRef] [PubMed]
- Djuric, Z.; Ellsworth, J.S.; Weldon, A.L.; Ren, J.; Richardson, C.R.; Resnicow, K.; Newman, L.A.; Hayes, D.F.; Sen, A. A Diet and Exercise Intervention during Chemotherapy for Breast Cancer. Open Obes. J. 2011, 3, 87–97. [Google Scholar] [CrossRef] [PubMed]
- Jacot, W.; Arnaud, A.; Jarlier, M.; Lefeuvre-Plesse, C.; Dalivoust, P.; Senesse, P.; Azzedine, A.; Tredan, O.; Sadot-Lebouvier, S.; Mas, S.; et al. Brief Hospital Supervision of Exercise and Diet During Adjuvant Breast Cancer Therapy Is Not Enough to Relieve Fatigue: A Multicenter Randomized Controlled Trial. Nutrients 2020, 12, 3081. [Google Scholar] [CrossRef]
- Maurer, T.; Belau, M.H.; von Grundherr, J.; Schlemmer, Z.; Patra, S.; Becher, H.; Schulz, K.H.; Zyriax, B.C.; Schmalfeldt, B.; Chang-Claude, J. Randomised controlled trial testing the feasibility of an exercise and nutrition intervention for patients with ovarian cancer during and after first-line chemotherapy (BENITA-study). BMJ Open 2022, 12, e054091. [Google Scholar] [CrossRef]
- Raghunath, K.; Sumathi, C.; Rajappa, S.J.; Mohan, M.; Kumar, U.; Shaik, U.; Botlagunta, M. Impact of naturopathy, yoga, and dietary interventions as adjuvant chemotherapy in the management of stage II and III adenocarcinoma of the colon. Int. J. Color. Dis. 2020, 35, 2309–2322. [Google Scholar] [CrossRef]
- Sanft, T.; Harrigan, M.; McGowan, C.; Cartmel, B.; Zupa, M.; Li, F.Y.; Ferrucci, L.M.; Puklin, L.; Cao, A.; Nguyen, T.H.; et al. Randomized Trial of Exercise and Nutrition on Chemotherapy Completion and Pathologic Complete Response in Women with Breast Cancer: The Lifestyle, Exercise, and Nutrition Early After Diagnosis Study. J. Clin. Oncol. 2023, 41, 5285–5295. [Google Scholar] [CrossRef] [PubMed]
- Stelten, S.; van Lonkhuijzen, L.; Hartman, Y.A.W.; van Driel, W.J.; Winkels, R.M.; Kenter, G.G.; Buffart, L.M.; Hoedjes, M. Experiences, adherence and satisfaction with a combined exercise and dietary intervention for patients with ovarian cancer undergoing chemotherapy: A mixed-methods study. Gynecol. Oncol. 2022, 165, 619–628. [Google Scholar] [CrossRef] [PubMed]
- Bille, S.J.; Fjalstad, B.W.; Clausen, M.B.; Andreasen, B.J.; Andersen, J.R. The Effect of Special Diets on Weight and Nutritional Intake in Hematological Cancer Patients: A Randomized Study. Nutr. Cancer 2018, 70, 874–878. [Google Scholar] [CrossRef] [PubMed]
- Gardner, A.; Mattiuzzi, G.; Faderl, S.; Borthakur, G.; Garcia-Manero, G.; Pierce, S.; Brandt, M.; Estey, E. Randomized comparison of cooked and noncooked diets in patients undergoing remission induction therapy for acute myeloid leukemia. J. Clin. Oncol. 2008, 26, 5684–5688. [Google Scholar] [CrossRef]
- IJmker-Hemink, V.; Lize, N.; Beijer, S.; Raijmakers, N.; Wanten, G.; van den Berg, M. Lessons learned from a randomized controlled trial on a home delivered meal service in advanced cancer patients undergoing chemotherapy: A pilot study. BMC Nutr. 2021, 7, 4. [Google Scholar] [CrossRef] [PubMed]
- IJmker-Hemink, V.E.; Kooijman, N.; Kamm, Y.J.L.; Koornstra, R.H.T.; Timmer-Bonte, J.N.H.; Wanten, G.J.A.; van den Berg, M.G.A. The Effect of a Home-delivered Protein-rich Meal Service on Quality of Life in Oncological Patients Undergoing Chemotherapy. Cancer Care Res. Online 2023, 3, e038. [Google Scholar] [CrossRef]
- Jalali, S.M.; Abdollahi, M.; Hosseini, A.; Bozorg, D.K.; Ajami; Azadeh, M.; Moiniafshar, K. The positive effects of Mediterranean-neutropenic diet on nutritional status of acute myeloid leukemia patients under chemotherapy. Front. Biol. 2018, 13, 475–480. [Google Scholar] [CrossRef]
- Kleckner, A.S.; Reschke, J.E.; Kleckner, I.R.; Magnuson, A.; Amitrano, A.M.; Culakova, E.; Shayne, M.; Netherby-Winslow, C.S.; Czap, S.; Janelsins, M.C.; et al. The Effects of a Mediterranean Diet Intervention on Cancer-Related Fatigue for Patients Undergoing Chemotherapy: A Pilot Randomized Controlled Trial. Cancers 2022, 14, 4202. [Google Scholar] [CrossRef] [PubMed]
- Sathiaraj, E.; Afshan, K.; R, S.; Jadoni, A.; Murugan, K.; Patil, S.; Naik, R. Effects of a Plant-Based High-Protein Diet on Fatigue in Breast Cancer Patients Undergoing Adjuvant Chemotherapy—A Randomized Controlled Trial. Nutr. Cancer 2023, 75, 846–856. [Google Scholar] [CrossRef] [PubMed]
- Villarini, A.; Pasanisi, P.; Raimondi, M.; Gargano, G.; Bruno, E.; Morelli, D.; Evangelista, A.; Curtosi, P.; Berrino, F. Preventing weight gain during adjuvant chemotherapy for breast cancer: A dietary intervention study. Breast Cancer Res. Treat. 2012, 135, 581–589. [Google Scholar] [CrossRef]
- Zhang, Y.; Cui, G.; Xiu, Y.; Zhao, M. The Effect of Nutritional Support Based on the Dietary Anti-Inflammatory Index on Cancer-Related Fatigue in Lung Cancer Patients Undergoing Chemotherapy. Cancer Nurs. 2023, 46, 394–404. [Google Scholar] [CrossRef] [PubMed]
- Cao, J.; Chen, C.; Wang, Y.; Liu, M.; Han, X.; Li, H. A nurse-led multidomain intervention to improve the management of chemotherapy-induced nausea and vomiting in patients with head and neck cancers: A randomized controlled trial. Eur. J. Oncol. Nurs. 2024, 70, 102615. [Google Scholar] [CrossRef]
- Ford, K.L.; Sawyer, M.B.; Ghosh, S.; Trottier, C.F.; Disi, I.R.; Easaw, J.; Mulder, K.; Koski, S.; Porter Starr, K.N.; Bales, C.W.; et al. Feasibility of two levels of protein intake in patients with colorectal cancer: Findings from the Protein Recommendation to Increase Muscle (PRIMe) randomized controlled pilot trial. ESMO Open 2024, 9, 103604. [Google Scholar] [CrossRef]
- Tang, F.; Wang, P.; Ye, Y. Effect of Nutritional Intervention on the Management of Radiotherapy and Chemotherapy for Nasopharyngeal Carcinoma. Nutr. Cancer 2024, 76, 114–120. [Google Scholar] [CrossRef] [PubMed]
- Brouwer, C.G.; Hartman, Y.A.W.; Stelten, S.; Kenkhuis, M.F.; van Lonkhuijzen, L.; Kenter, G.G.; Kos, M.; van de Ven, P.M.; Driel, W.J.V.; Winkels, R.M.; et al. Effects of a combined exercise and dietary intervention on clinical outcomes in patients with ovarian cancer: The Physical Activity and Dietary intervention in OVArian cancer (PADOVA) randomized controlled trial. Int. J. Gynecol. Cancer 2024, 1–7. [Google Scholar] [CrossRef]
- Kenkhuis, M.F.; Stelten, S.; Hartman, Y.A.; Brouwer, C.G.; Ten Tusscher, M.R.; van Lonkhuijzen, L.R.; Kenter, G.G.; van Driel, W.J.; Winkels, R.M.; Bekkers, R.L.; et al. Effects of a combined exercise and dietary intervention on body composition, physical functioning and fatigue in patients with ovarian cancer: Results of the PADOVA trial. Br. J. Cancer 2024, 131, 101–109. [Google Scholar] [CrossRef]
- Puklin, L.S.; Ferrucci, L.M.; Harrigan, M.; McGowan, C.; Zupa, M.; Cartmel, B.; Li, F.Y.; Ligibel, J.A.; Spiegelman, D.; Sharifi, M.; et al. Improving lifestyle behaviors during chemotherapy for breast cancer: The Lifestyle, Exercise, and Nutrition Early After Diagnosis (LEANer) Trial. Cancer 2024, 130, 2440–2452. [Google Scholar] [CrossRef] [PubMed]
- Krebs-Smith, S.M.; Pannucci, T.E.; Subar, A.F.; Kirkpatrick, S.I.; Lerman, J.L.; Tooze, J.A.; Wilson, M.M.; Reedy, J. Update of the Healthy Eating Index: HEI-2015. J. Acad. Nutr. Diet. 2018, 118, 1591–1602. [Google Scholar] [CrossRef] [PubMed]
- Silvers, M.A.; Savva, J.; Huggins, C.E.; Truby, H.; Haines, T. Potential benefits of early nutritional intervention in adults with upper gastrointestinal cancer: A pilot randomised trial. Support. Care Cancer 2014, 22, 3035–3044. [Google Scholar] [CrossRef]
- Djuric, Z. The Mediterranean diet: Effects on proteins that mediate fatty acid metabolism in the colon. Nutr. Rev. 2011, 69, 730–744. [Google Scholar] [CrossRef] [PubMed]
- Thompson, K.L.; Elliott, L.; Fuchs-Tarlovsky, V.; Levin, R.M.; Voss, A.C.; Piemonte, T. Oncology Evidence-Based Nutrition Practice Guideline for Adults. J. Acad. Nutr. Diet. 2017, 117, 297–310.e47. [Google Scholar] [CrossRef] [PubMed]
- Kleckner, A.S.; Magnuson, A. The nutritional needs of older cancer survivors. J. Geriatr. Oncol. 2022, 13, 738–741. [Google Scholar] [CrossRef] [PubMed]
- Mislang, A.R.; Di Donato, S.; Hubbard, J.; Krishna, L.; Mottino, G.; Bozzetti, F.; Biganzoli, L. Nutritional management of older adults with gastrointestinal cancers: An International Society of Geriatric Oncology (SIOG) review paper. J. Geriatr. Oncol. 2018, 9, 382–392. [Google Scholar] [CrossRef] [PubMed]
- Dieli-Conwright, C.M.; Wong, L.; Waliany, S.; Bernstein, L.; Salehian, B.; Mortimer, J.E. An observational study to examine changes in metabolic syndrome components in patients with breast cancer receiving neoadjuvant or adjuvant chemotherapy. Cancer 2016, 122, 2646–2653. [Google Scholar] [CrossRef] [PubMed]
- Slawinski, C.G.V.; Barriuso, J.; Guo, H.; Renehan, A.G. Obesity and Cancer Treatment Outcomes: Interpreting the Complex Evidence. Clin. Oncol. (R. Coll. Radiol.) 2020, 32, 591–608. [Google Scholar] [CrossRef]
- Hurria, A.; Lichtman, S.M.; Gardes, J.; Li, D.; Limaye, S.; Patil, S.; Zuckerman, E.; Tew, W.; Hamlin, P.; Abou-Alfa, G.K.; et al. Identifying vulnerable older adults with cancer: Integrating geriatric assessment into oncology practice. J. Am. Geriatr. Soc. 2007, 55, 1604–1608. [Google Scholar] [CrossRef] [PubMed]
- Chan, J.M.; Van Blarigan, E.L.; Langlais, C.S.; Zhao, S.; Ramsdill, J.W.; Daniel, K.; Macaire, G.; Wang, E.; Paich, K.; Kessler, E.R.; et al. Feasibility and Acceptability of a Remotely Delivered, Web-Based Behavioral Intervention for Men With Prostate Cancer: Four-Arm Randomized Controlled Pilot Trial. J. Med. Internet Res. 2020, 22, e19238. [Google Scholar] [CrossRef] [PubMed]
- Ludwig, D.S. The Ketogenic Diet: Evidence for Optimism but High-Quality Research Needed. J. Nutr. 2020, 150, 1354–1359. [Google Scholar] [CrossRef] [PubMed]
- Crosby, L.; Davis, B.; Joshi, S.; Jardine, M.; Paul, J.; Neola, M.; Barnard, N.D. Ketogenic Diets and Chronic Disease: Weighing the Benefits Against the Risks. Front. Nutr. 2021, 8, 702802. [Google Scholar] [CrossRef] [PubMed]
- Li, S.; Du, Y.; Meireles, C.; Sharma, K.; Qi, L.; Castillo, A.; Wang, J. Adherence to ketogenic diet in lifestyle interventions in adults with overweight or obesity and type 2 diabetes: A scoping review. Nutr. Diabetes 2023, 13, 16. [Google Scholar] [CrossRef]
- Campbell, E.K.; Fidahusain, M.; Campbell Ii, T.M. Evaluation of an Eight-Week Whole-Food Plant-Based Lifestyle Modification Program. Nutrients 2019, 11, 2068. [Google Scholar] [CrossRef]
- Santos, L.D.; Custodio, D.D.; Silva, A.T.F.; Ferreira, C.C.; Marinho, C.; Caixeta, C.; Souza, V.; Teixeira, R.; Araujo, T.G.; Shivappa, N.; et al. Overweight Women with Breast Cancer on Chemotherapy Have More Unfavorable Inflammatory and Oxidative Stress Profiles. Nutrients 2020, 12, 3303. [Google Scholar] [CrossRef]
- Groschel, C.; Prinz-Wohlgenannt, M.; Mesteri, I.; Karuthedom George, S.; Trawnicek, L.; Heiden, D.; Aggarwal, A.; Tennakoon, S.; Baumgartner, M.; Gasche, C.; et al. Switching to a Healthy Diet Prevents the Detrimental Effects of Western Diet in a Colitis-Associated Colorectal Cancer Model. Nutrients 2019, 12, 45. [Google Scholar] [CrossRef] [PubMed]
- Inglis, J.E.; Lin, P.J.; Kerns, S.L.; Kleckner, I.R.; Kleckner, A.S.; Castillo, D.A.; Mustian, K.M.; Peppone, L.J. Nutritional Interventions for Treating Cancer-Related Fatigue: A Qualitative Review. Nutr. Cancer 2019, 71, 21–40. [Google Scholar] [CrossRef] [PubMed]
- Mosher, C.E.; Champion, V.L.; Hanna, N.; Jalal, S.I.; Fakiris, A.J.; Birdas, T.J.; Okereke, I.C.; Kesler, K.A.; Einhorn, L.H.; Given, B.A.; et al. Support service use and interest in support services among distressed family caregivers of lung cancer patients. Psychooncology 2013, 22, 1549–1556. [Google Scholar] [CrossRef] [PubMed]
- Knippen, K.L.; Mahas, R.; Van Wasshenova, E. Outcome Expectancies, Health Information Seeking, and Cancer Beliefs Associated with Multivitamin/Mineral Use in a National Sample, HINTS-FDA 2015. J. Acad. Nutr. Diet. 2020, 120, 1368–1376. [Google Scholar] [CrossRef] [PubMed]
- Barak-Nahum, A.; Haim, L.B.; Ginzburg, K. When life gives you lemons: The effectiveness of culinary group intervention among cancer patients. Soc. Sci. Med. 2016, 166, 1–8. [Google Scholar] [CrossRef] [PubMed]
- Miller, M.F.; Li, Z.; Habedank, M. A Randomized Controlled Trial Testing the Effectiveness of Coping with Cancer in the Kitchen, a Nutrition Education Program for Cancer Survivors. Nutrients 2020, 12, 3144. [Google Scholar] [CrossRef]
- Wang, J.; Huang, Y. Pharmacogenomics of sex difference in chemotherapeutic toxicity. Curr. Drug Discov. Technol. 2007, 4, 59–68. [Google Scholar] [CrossRef] [PubMed]
- Siegel, R.L.; Miller, K.D.; Wagle, N.S.; Jemal, A. Cancer statistics, 2023. CA Cancer J. Clin. 2023, 73, 17–48. [Google Scholar] [CrossRef] [PubMed]
- Kim, H.I.; Lim, H.; Moon, A. Sex Differences in Cancer: Epidemiology, Genetics and Therapy. Biomol. Ther. 2018, 26, 335–342. [Google Scholar] [CrossRef]
- Robison, L.S.; Albert, N.M.; Camargo, L.A.; Anderson, B.M.; Salinero, A.E.; Riccio, D.A.; Abi-Ghanem, C.; Gannon, O.J.; Zuloaga, K.L. High-Fat Diet-Induced Obesity Causes Sex-Specific Deficits in Adult Hippocampal Neurogenesis in Mice. eNeuro 2020, 7, ENEURO.0391-19.2019. [Google Scholar] [CrossRef] [PubMed]
- Trujillo, E.B.; Claghorn, K.; Dixon, S.W.; Hill, E.B.; Braun, A.; Lipinski, E.; Platek, M.E.; Vergo, M.T.; Spees, C. Inadequate Nutrition Coverage in Outpatient Cancer Centers: Results of a National Survey. J. Oncol. 2019, 2019, 7462940. [Google Scholar] [CrossRef] [PubMed]
- Trujillo, E.B.; Dixon, S.W.; Claghorn, K.; Levin, R.M.; Mills, J.B.; Spees, C.K. Closing the Gap in Nutrition Care at Outpatient Cancer Centers: Ongoing Initiatives of the Oncology Nutrition Dietetic Practice Group. J. Acad. Nutr. Diet. 2018, 118, 749–760. [Google Scholar] [CrossRef] [PubMed]
- Sullivan, E.S.; Rice, N.; Kingston, E.; Kelly, A.; Reynolds, J.V.; Feighan, J.; Power, D.G.; Ryan, A.M. A national survey of oncology survivors examining nutrition attitudes, problems and behaviours, and access to dietetic care throughout the cancer journey. Clin. Nutr. ESPEN 2021, 41, 331–339. [Google Scholar] [CrossRef] [PubMed]
- Sa-Nguansai, S.; Pintasiri, P.; Tienchaiananda, P. Efficacy of oral nutritional supplement in cancer patients receiving chemotherapy: A systematic review and meta-analysis of randomized controlled trials. Ann. Palliat. Med. 2024, 13, 260–272. [Google Scholar] [CrossRef]
- Movahed, S.; Seilanian Toussi, M.; Pahlavani, N.; Motlagh, A.G.; Eslami, S.; Nematy, M.; Ghayour-Mobarhan, M.; Khadem-Rezaiyan, M.; Emadzadeh, M.; Varshoee Tabrizi, F.; et al. Effects of medical nutrition therapy compared with general nutritional advice on nutritional status and nutrition-related complications in esophageal cancer patients receiving concurrent chemoradiation: A randomized controlled trial. Mediterr. J. Nutr. Metab. 2020, 13, 265–276. [Google Scholar] [CrossRef]
Reference (Country) | Study Design | Sample Size | Eligibility | Intervention Type | Intervention Details | Primary Outcome(s) (Not Including Feasibility) |
---|---|---|---|---|---|---|
Nutrition counseling and/or food provision without exercise | ||||||
Abdollahi et al., 2019 (Iran) [19] | Randomized controlled trial | Randomized N = 150 | Female adults (≥18 y) with breast cancer who have received one session of chemotherapy (any type) and have a history of constipation or diarrhea after chemotherapy. | Nutrition counseling | Trial duration: 10-week trial. Intervention: Face-to-face nutrition education consisting of a diet with 12–15% calories of protein, 30–35% of fat, and 55–60% of carbohydrates estimated individually based on participant’s current age, weight, and height in addition to nutrition recommendations to reduce diarrhea and prevent reflux. Control: Control group had usual care without informational pamphlet, nutritional education, or dietary intervention, and they were asked not to change their usual diets. | Gastrointestinal side effects—nausea, vomiting, and diarrhea |
Baldwin et al., 2011 (United Kingdom) [20] | Randomized controlled trial | Randomized N = 277 | Adults with metastatic or locally advanced cancer of the gastrointestinal tract, lung, or mesothelioma on any type of chemotherapy; lost weight in the last 3 months; agreed to undergo and were fit for palliative chemotherapy | Nutrition counseling | Trial duration: 6 weeks, with a 52-week follow-up. Intervention 1: Advice to increase food intake by 600 kcal (2510 kJ) per day with the intent to increase body weight; booklet contained 150 kcal ideas and patients were asked to select 4/day. Intervention 2: A 588 kcal (2460 kJ) sachet of nutritional supplement to be prepared in whole milk + a daily multivitamin Control: No dietary intervention. | Survival |
Bourdel- Marchasson et al., 2014 (France) [21] | Randomized controlled trial | Randomized N = 341 | Patients older than 70 y with lymphoma or carcinoma with an indication of chemotherapy (any type); participants needed to be at risk of malnutrition with a full Mini Nutritional Assessment: 17–23.5 points | Nutrition counseling | Trial duration: 3 to 6 months, each participant received 6 visits. Intervention: Aim of achieving an energy intake of 30 kcal/kg body weight/d and 1.2 g protein/kg/d by face-to-face discussion targeting the main nutritional symptoms, compared to usual care. Control: Usual nutritional care given in cancer treatment settings. There were no restrictions for dietary advice or oral supplements. | One-year survival |
Cao et al., 2024 (China) [56] | Randomized controlled trial | Randomized N = 92 | Adults (≥18 y) diagnosed with head and neck squamous cell carcinoma, planned to receive first round of chemotherapy (cisplatin-based) | Nutrition counseling | Trial duration: 24 weeks. Intervention: Nurse-led multidomain intervention of chemotherapy-induced nausea and vomiting (CINV) included nurse-led risk assessment, education on prevention and control of CINV, antiemetic treatments following guidelines, dietary strategies (based on the National Comprehensive Cancer Network), relaxation therapy, and follow-up. Individualized dietary counseling was provided by the nurse tailored to patients’ specific needs. Control: Usual care of CINV consisting of administration of antiemetics, education about CINV control, and dietary recommendations provided by primary nurses. | Incidence of CINV, severity of CINV, influence of CINV on patients’ quality of life |
Dai et al., 2022 (China) [22] | Pilot randomized controlled trial | Randomized N = 72 | Men and women aged 18–75 y, head or neck cancer, concurrent chemoradiation therapy (nedaplatin) | Nutrition counseling | Trial duration: Duration of the concurrent chemoradiation therapy. Intervention: Face-to-face nutritional counseling at least once every 2 weeks during treatment to achieve calorie and protein requirements determined by the ESPEN guidelines on nutrition in cancer patients. Control: Nutrition treatment plans according to clinical experience. | Body weight |
de Lima Bezerra et al., 2023 (Brazil) [23] | Randomized controlled trial | Randomized N = 34 | Adult women with breast cancer, indicated for neoadjuvant chemotherapy (any type) | Nutrition counseling | Trial duration: 2 months. Intervention: Personalized diet with an individually tailored meal plan based on ESPEN guidelines to address malnutrition [9] (25–30 kcal/kg body weight/day and 1.5 g protein/kg/day). Control: Counseling on healthy eating and nutritional information on how to reduce the severity of chemotherapy-induced nausea and vomiting. | Chemotherapy toxicity |
de Souza et al., 2021 (Brazil) [24] | Randomized controlled trial | Randomized N = 34 | Female adults with breast cancer indicated for neoadjuvant chemotherapy (doxorubicin and cyclophosphamide) | Nutrition counseling | Trial duration: 3 cycles of chemotherapy (21 days/cycle). Intervention: A personalized diet meal plan formulated by a dietitian based on age, current weight, and height of each subject, 25–30 kcal/kg/day of energy and 1.5 g/kg/day of protein, based on the ESPEN guidelines [9], including written materials on healthy eating and symptom management Control: Pamphlets with recommendations on healthy eating and to reduce chemotherapy-induced nausea and vomiting. | Quality of life |
Ford et al., 2024 (Canada) [57] | Randomized controlled pilot trial | Randomized N = 50 | Adults (18–85 years) with stage II–IV colorectal cancer starting any type of chemotherapy | Nutrition counseling | Trial duration: 12 weeks. Intervention groups: Weekly telephone nutrition education with patients to achieve 2.0 g protein/kg body weight/day and meal patterns consistent with those recommended by the Academy of Nutrition and Dietetics. Control: Weekly telephone nutrition education with patients to achieve a protein intake of 1.0 g/kg/day (standard of care). Both: Prescribed diets were translated into an individualized daily meal pattern based on typical dietary intake. | Muscle mass and physical function |
Keum et al., 2021 (Korea) [25] | Randomized controlled trial | Randomized N = 40 | Males or females aged 20–70 y with pancreatic ductal adenocarcinoma scheduled to receive chemotherapy (any type) | Nutrition counseling via a smartphone app | Trial duration: 12 weeks. Intervention: Noom is a mobile app that offers a curriculum and human coaching. It was tailored to the population and included food logging, feedback-based food choices, daily articles for health, and coach–participant messaging with goals to encourage calorie intake and maintain nutritional status. Control: Usual care. | Nutritional status and quality of life |
Lin et al., 2017 (China) [26] | Randomized controlled trial | Randomized N = 110 | Males and females aged ≥18 y with cancer (any type) and undergoing chemotherapy (any type) | Nutrition counseling | Trial duration: 12 cycles of chemotherapy (duration in days not specified). Intervention: Individual recipes based on estimates of total energy expenditure to achieve or maintain high nutritional status. Participants and their caretakers were counseled by experienced nurses, and diets were adjusted as nutritional status changed throughout the intervention. Control: Usual care plus nutrition screening, nutritional guidance, diet mission, and arranged mealtimes. | Patient weight and serum albumin and prealbumin levels |
Loprinzi et al., 1996 (United States) [27] | Randomized controlled trial | Randomized N = 109 | Premenopausal women with resected localized breast cancer scheduled to receive adjuvant chemotherapy (any type) | Nutrition counseling | Trial duration: Duration of chemotherapy, assessments at 3 and 6 months after initiation of chemotherapy. Intervention: Counseling with a dietitian prior to or within 2 weeks of chemotherapy initiation and at 4-to 6-week intervals. Dietitian goals included maintaining on-study body weight or gradual weight reduction toward ideal body weight. Control: Standard care—information on the potential for weight gain and ways to prevent it. | Body weight |
Najafi et al., 2019 (Iran) [28] | Same as Abdollahi et al. [19] | Randomized N = 150 | Females aged 18–60 y with stage IA–IIIB breast cancer being treated with thrice-weekly cycles of doxorubicin, cyclophosphamide, and docetaxel and had experienced vomiting during or after the previous session(s) of chemotherapy | Nutrition counseling | Trial duration: Duration of chemotherapy. Intervention: Personalized diet and nutrition education before each chemotherapy session. The daily diet was estimated individually and contained 1.2–1.5 g/kg of protein, 30% of energy from fat, and 55–60% of energy from carbohydrates. Control: Usual care. | Chemotherapy-induced nausea and vomiting |
Ollenschläger et al. (Germany) 1992 [29] | Randomized controlled trial | Enrolled N = 32 | Males and females aged 17–60 years with acute lymphocytic or nonlymphocytic leukemia; had unintentional weight loss of >5% in the previous 3 months and/or actual body weight below 90% ideal body weight; planned cytostatic treatment with lamivudine-6; thioguanine, Ara-C, and daunorubicin (TAD-9); or the Ulm protocol | Nutrition counseling | Trial duration: Duration of therapy from induction of therapy until planned or premature end of maintenance therapy (4–54 weeks); participants were hospitalized for some of their cancer treatment and some were on an out-patient basis. Intervention: “Intensified oral nutrition program”: ad libitum menu of 1.0–2.0 g protein and 30–50 kcal/kg body weight plus daily visit by a dietitian to assess nutritional status, nutritional behavior, nutrient requirements, symptoms, and well-being. Control: Free choice, ad libitum menu of 1.0–2.0 g protein and 30–50 kcal/kg body weight. | Quality of life |
Ovesen et al., 1993 (Denmark) [30] | Randomized controlled trial | Enrolled N = 137 | Males and females with small-cell lung cancer, ovarian cancer, or breast cancer on any type of chemotherapy | Nutrition counseling | Trial duration: 5 months. Intervention: Counseling with a dietitian before chemotherapy and once–twice monthly and as needed; goals to meet the protein and calorie requirements of the Nordic Recommended Allowances (1.5–1.7 × basal energy expenditure as calculated by the Harris–Benedict Equation; 1.0–1.2 g protein/kg body weight) Control: Ad libitum diet. Both groups: Advised to supplement diet with one multivitamin tablet that contained the recommended daily allowances of micronutrients. | Body weight, quality of life |
Regueme et al., 2021 (France) [31] | Same as Bourdel-Marchasson et al. [21] | Randomized N = 283 | Health-related quality of life | |||
Sukaraphat et al., 2016 (Thailand) [32] | Randomized controlled study | Enrolled N = 50 | Adults (age ≥ 18 years) with locally advanced unresectable or metastatic cancer on first-line chemotherapy (any type) | Nutrition counseling | Trial duration: 9–12 weeks (3–4 cycles of chemotherapy). Intervention: Individualized and intensive dietary counseling based on the ESPEN guideline [3]: 30–35 kcal/kg body weight and 0.8–1.2 g protein/kg body weight per day with regular food, supplements used if required. Control: Routine care: dietary counseling for general dietary recommendations from a nurse and physician but not a dietitian. | Body weight |
Tang et al., 2024 (China) [58] | Quasi-experimental study | Randomized N = 60 | Adults (≥18 y) with nasopharyngeal carcinoma who were on this type of chemotherapy (any type) and had been diagnosed with malnutrition (weight loss ≥ 5%) and had a life expectancy of more than 3 months. | Nutrition counseling | Trial duration: 24 weeks. Intervention: Face-to-face nutrition education with patients and their families consisting of a diet high in protein, high in calories, high in vitamins, and rich in inorganic salts, in addition to nutrition recommendations to avoid dry mouth, oropharyngeal pain, and constipation consistent with guidelines. Control: Routine dietary guidance without nutrition assessment screening. | Nutritional Risk Screening 2002, BMI, and quality of life |
van der Werf et al., 2020 (Netherlands) [33] | Randomized controlled trial | Randomized N = 107 | Males and females aged > 18 y diagnosed with metastatic colorectal cancer, scheduled for first-line chemotherapy with capecitabine and oxaliplatin (CAPOX), infusional 5- fluorouracil and oxaliplatin (FOLFOX) or capecitabine alone, with or without bevacizumab | Nutrition counseling | Trial duration: Duration of chemotherapy (varied by participant based on regimen). Intervention: Face-to-face counseling (at every chemotherapy infusion) and telephone consultation (between infusions) by a dietitian with the goal of achieving sufficient protein, energy, and micronutrient intake. The protein intake goal was ≥1.2 g protein/kg body weight/day. The dietitian also encouraged moderate-intensity physical activity for ≥30 min per day, five days per week. Control: Access to a dietitian. | Skeletal muscle area (primary), quality of life, physical functioning, treatment toxicity, treatment intensity, survival (secondary) |
Wang et al., 2023 (China) [34] | Randomized control trial | Randomized N = 53 | Males and females aged 50–80 years with esophageal cancer undergoing chemoradiotherapy (cisplatin + paclitaxel or cisplatin + 5-fluorouracil + radiation) | Nutrition counseling | Trial duration: Duration of chemoradiotherapy (5–8 weeks). Intervention: Individualized nutrition programs with target energy and protein intake (30–35 kcal/kg body weight/day and 1.2–1.5 g protein/kg/day). Nutritional evaluations were conducted by nutrition nurses each day, and nutritional interventions were adjusted based on needs. Control: Usual care. | Nutritional status |
White et al., 2020 (United Kingdom) [35] | Randomized control trial | Randomized N = 50 | Males and females aged > 18 y, with cervical or bladder cancer, undergoing chemotherapy and radical pelvic radiotherapy (weekly gemcitabine for bladder cancer or cisplatin for cervical cancer) | Nutrition counseling | Trial duration: Not specified, 6-week, 12-week, and 12-month follow-up. Intervention: Individualized dietary counseling using the anthropometric data, patient-reported subjective, Global Assessment score, Malnutrition Universal Screening Tool score, dietician symptom assessment, and 3-day food diary to tailor dietary advice and prescription of oral nutritional supplements where necessary. Control: Not stated. | Feasibility (primary), treatment toxicity (primary clinical outcome) |
Xie et al., 2017 (China) [36] | Randomized control trial | Randomized N = 144 | Males and females aged ≥ 18 y with gastric cancer undergoing post-operative chemotherapy (oxaliplatin and capecitabine every 21 days for up to 8 cycles) | Nutrition counseling | Trial duration: Duration of chemotherapy. Intervention: Intensive individualized nutritional and educational interventions during the entire course of chemotherapy. Control: Education on chemotherapy, possible chemotherapy-induced adverse events, basic nutrition guidance, follow-up 2 days after infusion. | Nutritional status |
Nutrition counseling plus exercise | ||||||
Allen et al., 2022 (United Kingdom) [37] | Pilot randomized controlled trial | Randomized N = 54 | Adults with locally advanced esophagogastric cancer planned for neoadjuvant therapy (any type) plus esophagogastrectomy, or total gastrectomy. | Nutrition counseling and exercise | Trial duration: 15-week trial (preoperative). Intervention: Needs-based, frequent, and tailored to the patient by dietitians to increase total calories and protein where appropriate. This group also received a structured exercise program. Control: Standard care, no nutrition or exercise prehabilitation. | Cardiopulmonary exercise test performance, muscle mass, and quality of life |
Basen-Engquist et al., 2020 (United States) [38] | Pilot randomized controlled trial | Randomized N = 39 | Patients with newly diagnosed stage II or III breast cancer within 3 weeks of starting neoadjuvant chemotherapy (any type) | Nutrition counseling and exercise | Trial duration: ~6 month trial (until the end of chemotherapy). Intervention: 20 individualized counseling sessions, 14 in person and 6 by telephone. Promoted foods with low energy density and consistent with recommendations for survivors. Also, an exercise intervention focused on resistance and flexibility training to maintain or increase fat-free mass. Control: Provision of publicly available written materials on cancer survivorship. | Anthropometrics (to prevent weight gain), quality of life, serum biomarkers |
Brouwer et al., 2024 (Netherlands) [59] | Randomized controlled trial | Randomized N = 81 | Patients aged > 18 years with primary epithelial ovarian cancer who were scheduled for (neo)-adjuvant chemotherapy treatment (six 3-week cycles of adjuvant carboplatin with or without paclitaxel for early FIGO stage and adjuvant or neo-adjuvant carboplatin or cisplatin and paclitaxel for advanced FIGO stage). | Nutrition counseling and exercise | Trial duration: From first or second chemotherapy cycle until 12 weeks after the last chemotherapy cycle. Intervention: Two 1 h sessions per week, supervised by a physical therapist specialized in oncology, including moderate- to high-intensity resistance and aerobic exercise. Dietary counseling was delivered by dietitians specialized in oncology once every 3 weeks during 30–45 min face-to-face sessions in the hospital or by telephone tailored to the nutritional needs of each patient. All participants were advised to follow the dietary guidelines set by the World Cancer Research Fund (WCRF)/American Institute for Cancer Research (AICR) and were advised to consume at least 1.2 g of protein/kg body weight/day and at least 25 g of protein per meal. Control: Usual care during chemotherapy, which could include a referral to a dietitian when malnutrition was detected by the gynecological oncologist. | Relative dose intensity and progression-free survival |
Carayol et al., 2019 (France) [39] | Randomized controlled trial | Randomized N = 143 | Women aged 18–75 y; non-metastatic breast cancer; enrolled after curative surgery, planned for 6 cycles of adjuvant chemotherapy (epirubicin/cyclophosphamide/5-fluorouracil for 3 cycles every 3 weeks, followed by docetaxel for 3 cycles every 3 weeks), followed by 6 weeks of radiotherapy | Nutrition counseling and exercise | Trial duration: 26 weeks. Intervention: The Adapted Physical Activity and Diet (APAD) intervention consisted of exercise sessions planned thrice-weekly, nine hospital-based and face-to-face nutritional therapeutic education sessions targeting body weight control and feeding behaviors according to the WCRF. Specific advice was given to patients for the management of chemotherapy toxicities and side effects. Control: Usual care; no diet or exercise advice. | Cancer-related fatigue |
Demark- Wahnefried et al., 2008 (United States) [40] | 3-Arm randomized controlled trial | Randomized N = 90 | Women with stage I–IIIA breast cancer scheduled for adjuvant chemotherapy (any type) | Calcium-rich diet ± exercise ± high fruit and vegetable, low fat | Trial duration: 6 months All groups: 14 contacts of 10–30 min each, once weekly for the first month and semiweekly for 5 months Intervention 1 (calcium-rich diet + exercise): Calcium-rich intervention + aerobic exercise ≥30 min per day, ≥3 times weekly and strength training exercises every other day. Intervention 2 (calcium-rich, low-fat, high-fruit-and-vegetable diet + exercise): Intervention 1 + goals of <20% energy from fat, ≥5 servings fruit + vegetables. Control (calcium-rich diet only): Written and verbal instruction on a calcium-rich diet (1200–1500 mg calcium/day). | Body composition (preservation of lean mass) |
Djuric et al., 2011 (United States) [41] | Randomized controlled trial | Randomized N = 40 | Females age ≥18 y with stage I–IIIA breast cancer scheduled for or within two weeks of starting chemotherapy (any type) | Nutrition counseling and exercise | Trial duration: 12 months. Intervention: Written educational materials and telephone counseling from a registered dietitian to achieve a high-fruit-and-vegetable and low-fat diet, based on an estimate of total energy expenditure (TEE). Control: “My Pyramid” plan from the United States Department of Agriculture and brochures from the American Cancer Society. | Feasibility |
Jacot et al., 2020 (France) [42] | Randomized controlled trial | Randomized N = 360 Analyzed in a per-protocol analysis N = 321 | Females ≥ 18 y with early-stage breast cancer, participants were enrolled after undergoing curative surgery and before 6 cycles of chemotherapy (six cycles of adjuvant chemotherapy [three cycles of epirubicin/cyclophosphamide/5-fluorouracil every 3 weeks, followed by three taxane-based cycles, either docetaxel every 3 weeks or paclitaxel weekly for 9 weeks]) followed by 6 weeks of radiotherapy. | Nutrition counseling and exercise | Trial duration: 26 weeks. Intervention: The Adapted Physical Activity Diet (APAD2) program: twice weekly exercise sessions and six nutrition sessions, which targeted body weight control and adherence to WCRF recommendations. Control: Usual care. | Cancer-related fatigue |
Kenkhuis et al., 2024 (Netherlands) [60] | Same as Brouwer et al. [59] | Body composition, physical functioning, and fatigue | ||||
Maurer et al., 2022 (Germany) [43] | Randomized controlled trial | Randomized N = 15 Analyzed N = 11 | Females aged ≥ 18 y with ovarian cancer, tubal cancer, or peritoneal cancer, scheduled for adjuvant or neoadjuvant chemotherapy (any type) | Nutrition counseling and exercise | Trial duration: 12 months. Intervention: Personalized exercise and nutrition counselling tailored to different phases of the patient’s treatment and recovery. The nutrition element included one-on-one sessions with a nutritionist every 3 weeks during chemotherapy, with a focus on consuming adequate calories and protein. After chemotherapy, monthly nutrition counselling was focused on the Mediterranean diet. Control: Usual care. | Feasibility (primary); secondary: quality of life, cancer-related fatigue, nutritional risk, physical activity, physical performance, body composition |
Puklin et al., 2024 (United States) [61] | Randomized controlled trial | Randomized N = 173 | Women with stage I–III breast cancer receiving chemotherapy (any type) | Nutrition counseling and exercise | Trial duration: Duration of chemotherapy (average 3.3 ± 1.2 months, range 8–24 weeks). Intervention: Goals to meet guidelines according to the Healthy Eating Index-2015 [62] (≥5 fruits and/or vegetables/day, ≥25 g fiber/day, <30 g added sugar/day, ≤18 oz red meat/week, limited consumption of processed foods, ≤1 alcoholic drink/day) and Physical Activity Guidelines (≥150 min/week of moderate- to vigorous-intensity physical activity or 75 min/week of vigorous-intensity physical activity plus twice-weekly resistance training); a mean of 8 ± 3 counseling sessions were offered. Control: Usual care; access to a registered dietitian with referrals at the discretion of the treating oncologist. | Physical activity and diet quality |
Raghunath et al., 2020 (India) [44] | Randomized controlled trial | Randomized N = 120 | Males and females 18–65 y with stage II and III adenocarcinoma of the colon and prescribed chemotherapy (8 cycles with oxaliplatin and capecitabine for 14 days then 3 weeks off) | Naturopathy, yoga, and dietary counseling | Trial duration: 18 months. Intervention: The dietary intervention included a schedule of foods such as vegetable juice, idlis, korralu, dalia, and milk malt with buttermilk. Also, after the first cycle of chemotherapy, participants underwent a 7-day intensive phase of training in naturopathy and yoga. Control: Psycho-social counseling. | Hematological, biochemical, immunological, and psychological variables |
Sanft et al., 2023 (United States) [45] | Same as Puklin et al. [61] | Relative dose intensity (measure of chemotherapy completion) | ||||
Stelten et al., 2022 (Netherlands) [46] | Randomized controlled trial | Randomized N = 28 | Females aged ≥18 y with primary epithelial ovarian cancer scheduled to undergo first-line neoadjuvant chemotherapy (any type) | Nutrition counseling and exercise | Trial duration: Duration of chemotherapy plus follow-up 3 and 12 weeks after chemotherapy. Intervention: The dietary component involved individually tailored diets delivered via face-to-face sessions once every three weeks to achieve/maintain nutritional status and dietary intake and to prevent weight loss, guided by the WCRF/AICR (≥1.2 g protein/kg body weight/day evenly distributed throughout meals). The exercise component included two one-hour moderate to high-intensity resistance and aerobic exercise sessions per week by a physical therapist. Control: Usual care. | Body composition, physical function, and fatigue; though only qualitative data are reported in this paper. |
Nutrient-specific dietary patterns | ||||||
Bille et al., 2018 (Denmark) [47] This manuscript also reports two pilot studies (n = 5 each) to select meals for the cross-over study. Details from the pilot studies are not described here. | Randomized cross-over controlled trial | Enrolled N = 62 | Adults with hematological cancer being treated with chemotherapy (any type) | High energy density | Trial duration: 30-day control period followed by a 30-day intervention period. Intervention: Four high-energy-density dishes with protein content of at least 5 g/100 g each eaten once each: chili con carne, chicken in curry, curry soup, and pasta carbonara. Control: Habitual diet. | Acceptability of dishes, weight loss |
Harvey et al., 2023 (United States) [6] | Same as Kleckner et al. [52] | Qualitative assessment of participants’ experiences | ||||
Gardner et al., 2008 (United States) [48] | Randomized controlled trial | Randomized N = 153 | Males and females with acute myeloid leukemia receiving remission induction therapy | Neutropenic | Trial duration: Length of stay, 24–25 days. Intervention: No raw fruits or vegetables, only cooked. Control: A diet containing fresh fruit and fresh vegetables with encouraged consumption of at least one/day, with the fruits and vegetables washed with cold water for 30 s before being eaten. | Infection and death |
IJmker-Hemink et al., 2021 (Netherlands) [49] | Randomized controlled trial | Randomized N = 20 | Males and females aged ≥18 y with metastatic colorectal or gynecological malignancies starting with 3-weekly scheduled chemotherapy (any type) | Protein-rich home delivery | Intervention duration: 3 weeks (with follow-up for 3 months). Intervention: Home-delivered meal service of six protein-rich dishes per day based on the FoodforCare meal service. The meal service provided a morning shake, two lunch meals, a snack, dinner, and dessert for each day (average energy 1553 kcal/day, average protein 60.8 g/day) for 3 weeks. Participants received a leaflet with nutritional advice on protein-rich foods and recommendations for 1.2 g protein/kg body weight/day. Control: Usual care. | Quality of life |
IJmker-Hemink et al., 2023 (Netherlands) [50] | Randomized controlled trial | Randomized N = 148 | Males and females aged ≥18 y with cancer (any type) and receiving chemotherapy on a 2-, 3-, or 4-week cycle (any type) | Protein-rich home delivery | Same as IJmker-Hemink et al. [49] | Quality of life |
Jalali et al., 2018 (Iran) [51] | Randomized controlled trial | Randomized N = 50 | Males and females with acute myeloid leukemia undergoing chemotherapy (type not specified) | Mediterranean diet | Trial duration: 4 weeks. Intervention: Mediterranean diet with 47% carbohydrates, 15% protein, 38% fat (24% monounsaturated fatty acids mostly from olive oil), including 30 mL olive oil daily. Control: Neutropenic diet: cooked foods and vegetables, except for some raw fruits, including banana and orange; boiled water; pasteurized dairy; well cooked meat and eggs Both diets had a similar calorie count; the Mifflin formula was used to estimate energy needs. | Nutritional status |
Kleckner et al., 2022 (United States) [52] | Randomized controlled trial | Randomized N = 33 | Males and females aged ≥18 y with cancer (any type) with at least six weeks of chemotherapy remaining (any type) | Mediterranean diet | Trial duration: 8 weeks. Intervention: Participants received 12 frozen meals/week delivered to their homes once/week for four weeks and other grocery store items, a custom Mediterranean cookbook, and information describing the MedDiet. Participants had weekly phone-based check-ins with the study team and a one-on-one counseling session with a nutrition scientist during week 3. Control: Usual care. | Cancer-related fatigue |
Sathiaraj et al., 2023 (India) [53] | Randomized controlled trial | Randomized N = 103 | Females aged 18–65 years with nonmetastatic breast cancer and planning to have adjuvant chemotherapy (4 cycles of doxorubicin and cyclophosphamide or docetaxel and cyclophosphamide (1 cycle every 3 weeks) | Plant-based high protein | Trial duration: Duration of chemotherapy (about 12 weeks). Intervention: A plant-based, high-protein diet (1.2 g protein/kg body weight/day; 25–30% kcal from fat, 55–60% carbohydrates, 10–15% protein) aimed at weight management; advised no meat, included eggs and low-fat dairy, ≥5 servings fruits and vegetables, whole grain and millet, discouraged high-calorie fried food, soda; whey protein supplement (84 kcal, 10 g protein) once daily; educational materials; and weekly sessions/reviews. Control: Usual care, with access to on-demand nutrition appointments with the study team. | Cancer-related fatigue (primary), body composition (secondary) |
Villarini et al., 2012 (Italy) [54] | Randomized controlled trial | Randomized N = 96 | Women of any age, operated on for invasive, non-metastatic breast cancer, scheduled for adjuvant chemotherapy (any type) | Diet based on Mediterranean and macrobiotic recipes | Trial duration: Duration of chemotherapy. Intervention: Cooking classes and common meals at least twice per week in addition to the recommendations and kitchen course given to the control group; dietary recommendations based on Mediterranean and macrobiotic recipes and on the avoidance of energy-dense foods. Protein intake was reduced, as meat and cheese consumption was discouraged. Control: Recommendations for the prevention of cancer and a baseline kitchen course on how to reduce gastrointestinal side effects of chemotherapy. | Body weight |
Zhang et al., 2023 (China) [55] | Randomized controlled trial | Enrolled N = 106 | Patients with lung cancer undergoing chemotherapy (type not specified), body mass index 18.5–30 kg/m2, Brief Fatigue Inventory score of ≥4 points (range 0–10). | Anti-inflammatory diet | Trial duration: 3 months. Intervention: The anti-inflammatory diet goals: 2 servings of fruit, 5 servings of vegetables, 3 servings of whole grain cereals, 2 servings of nuts or seeds, 2 servings of olive oil or linseed oil, and 2 servings of probiotic low-fat dairy products per day; ≥3 servings of fish, ≤2 servings of red meat, and 5 servings of white meat per week; encouragement of certain spices and tea, discouragement of refined and processed foods; nutrition education; twice/week phone calls or face-to-face consultations. Control: Usual diet; 4 face-to-face visits for regular health education and recommendations based on dietary nutrition prescriptions for patients with cancer in China. | Cancer-related fatigue |
Reference | Sample Characteristics | Feasibility (Completion, Attendance, Compliance) | Safety (Adverse Events, Side Effects) | Efficacy |
---|---|---|---|---|
Nutritional counseling without exercise | ||||
Abdollahi et al., 2019 [19] | Analyzed N: 140 (73 intervention, 67 control) Sex: all female Mean age: 46.8 years Cancer type(s): breast | - 93.3% retention; 10/150 did not complete the study. - Compliance with the nutrition recommendations was >90% for the treatment group (details not described). | - None reported related to the intervention | - There were significant within-group decreases in the prevalence of gastrointestinal side effects in the intervention group in the third session of chemotherapy compared to the first session, including reflux disorder (p = 0.05), anorexia (p < 0.001), nausea (p = 0.002), constipation (p < 0.001), and diarrhea (p < 0.001); symptom prevalence was stable in the control group. - All 9 measured gastrointestinal side effects were lower in the intervention group vs. the control group at the fourth chemotherapy session, controlling for baseline presence and other potential confounding factors. |
Baldwin et al., 2011 [20] | Analyzed N: 358 (90 diet, 86 supplement, 86 diet + supplement, 96 control) Sex: 256 males, 102 females Median age: 66.8 (range 24–88 years) Cancer type(s): 113 colorectal, 81 lung, 72 pancreas, 71 esophago-gastric, 10 liver and biliary, 7 unknown primary, 4 other | - 326/358 (91.1%) retention at week 6 (primary endpoint), 153/358 (42.7%) retention at week 52; data available for 98% because most lost-to-follow-ups were because of death. - Only 60/236 (25%) completed a food diary at baseline and 40 (17%) completed it at more than one time point (data loss due mostly to deteriorating health status and death); therefore, analysis was deemed not useful. - Supplement intake logs were returned by 62/150 (41%) participants; 31% took all prescribed supplements the first week and 19% took them all at week 6; 5 (8%) were unable to take any supplements the first week and 48% were unable to take any supplements by week 6 | - No side effects reported that were attributed to the intervention | - One-year survival was 38.6% (95% confidence interval 33.3–43.9%), with no differences between groups (±dietary advice, ±supplement). - Quality of life, as measured using both the EORTC-C30 and the FAACT, were not different between the groups. - There were no between-group differences in weight change. - There were no between-group differences in hand grip strength. |
Bourdel-Marchasson et al., 2014 [21] | Analyzed N: 336 (169 intervention, 167 control) Sex: 172 males, 164 females Mean age: 78.0 years Cancer type(s): colon (49), stomach (25), pancreas and cholangiocarcinoma (62), non-small cell lung (35), prostate (13), bladder (20), ovary (25), breast (28), lymphoma (50) | - 98.5% retention. - 877/990 (88.6%) dietitian visits were attended. - In both groups, energy intake increased between visit 1 and visit 2 (intervention: +328 kcal/day, p < 0.0001; control: +132 kcal/day, p = 0.02) but the difference was higher in the intervention vs. control group (p < 0.01). - At visit 2, 57 (40.4%) participants in the intervention group vs. 13 (13.5%) in the control group achieved the goal of 30 kcal/kg/d or more and 66 (46.8%) in the intervention group vs. 20 (20.8%) in the control group achieved the goal of 1.2 g protein/kg/d. | - None reported related to the intervention | - One-year and two-year mortality were similar in both groups. - Clinical outcomes of chemotherapy management, remission status, weight change, serum albumin, incidents of falls, hospitalization, and use of artificial nutrition were similar in both groups (p ≥ 0.19). - There were more grade 3–4 infections among control vs. intervention participants (p = 0.03). |
Cao et al., 2024 (China) [56] | Analyzed N: 92 (45 intervention, 47 control) Sex: 77 males, 15 females Mean age: 61.1 years Cancer type(s): head and neck squamous cell carcinomas | - 92/94 (97.9%) completed the study. Two patients in the intervention group dropped out because chemotherapy was stopped. | - None reported related to the intervention | - Compared to the control group, patients in the intervention group had a significantly lower incidence of nausea (p = 0.021) and a significantly lower rate of vomiting (p = 0.035). - Regarding the patient-reported severity of nausea and vomiting, the intervention group showed better results compared to the control group (p = 0.014 and p = 0.029, respectively). - The scores of the nausea and vomiting domains of the Functional Living Index-Emesis in the intervention group were significantly higher than those in the control group (p < 0.001 and p = 0.018, respectively), indicating that patients in the intervention group perceived less impact of CINV on their daily lives. |
Dai et al., 2022 [22] | Analyzed N: 61 (32 intervention, 29 control) Sex: 50 males, 11 females Mean age: 51.7 years Cancer type(s): nasopharynx (51), hypo-pharynx (3), tonsil (1), cervical lymph node (1), tongue (1), larynx (3), oropharynx (1) | - 84.7% retention; 11/72 (15.3%) did not complete the study. - The decrease in calorie intake was attenuated in the intervention group vs. control (not significant at week 2, p < 0.001 at weeks 4 and 6). - Protein intake increased in the intervention group and decreased in the control group (p < 0.001 between groups at all 3 time points). | - None reported related to the intervention | - Body weight was higher in the intervention vs. control group (p = 0.03). - Karnofsky Performance Status score was higher in the intervention vs. control groups across all time points (p = 0.01). - Decreases in nutritional indexes albumin, transferrin, and pre-albumin, and triceps skin folds were higher in the control vs. intervention group (p < 0.05). - Anxiety and depression were higher in the control vs. intervention group over all time points (p ≤ 0.01). - Participants in both groups suffered from mucositis to different degrees. |
de Lima Bezerra et al., 2023 [23] | Analyzed N: 34 (19 intervention, 15 control) Sex: all females Mean age: 44–45 years Cancer type(s): breast | - 100% retention; no loss to follow-up. | - None reported related to the intervention | - There was less chemotherapy-induced gastrointestinal chemotoxicity in the intervention vs. control group for nausea (p < 0.001), vomiting (p = 0.048), and constipation (p = 0.033) but not diarrhea, anorexia, or mucositis. - The intervention group had a lower frequency of chemotherapy dose-limiting toxicity compared to the control group (15.8% vs. 26.7%, p = 0.672). - Changes in body weight were similar between groups. - There were no between-group differences in disease-free survival. |
de Souza et al., 2021 [24] | Analyzed N: 34 (19 intervention, 15 control) Sex: all females Mean age: 44.3 years Cancer type(s): breast | - 30/34 (88.2%) completed the study. | - None reported related to the intervention | - Global health status quality of life increased slightly over time for both groups, with no between-group differences. - Physical, emotional, cognitive, and social functioning were stable over time for both groups. - Role functioning was maintained in the intervention group and decreased over time for the control group (p < 0.001 between groups). - Anthropometrics (body weight, BMI, and calf circumference) decreased slightly over time for both groups to a similar degree (p > 0.1 over time for both groups). - Hand-grip strength decreased significantly over time for the control group (p = 0.009), and the decrease was less in the intervention group (p = 0.125 over time). - Nausea and vomiting scores were higher in the intervention group vs. the control group (p < 0.001 group × time); appetite loss decreased over time for the intervention group and remained stable for the control group (but was higher at baseline in the intervention group); there were no other effects of group or group × time for other quality-of-life symptoms. - In regard to hematological and gastrointestinal toxicities, the intervention group had a lower incidence of leukopenia in the third cycle of chemotherapy (p = 0.034) and a lower amount of abdominal pain in the second cycle (p = 0.034); no other between-group differences. |
Ford et al., 2024 (Canada) [57] | Analyzed N: 50 (25 intervention, 25 control) Sex: 30 males, 20 females Mean age: 57 years Cancer type(s): colorectal cancer | - 40/50 (80%) participants were retained (23/25 control and 17/25 intervention). - Both groups increased protein consumption from baseline to weeks 6 and 12, with those in the 2.0 g/kg group consuming more at week 6 and week 12. Being in the 2.0 g/kg/day group resulted in 0.3 g/kg/day greater protein intake compared with the 1.0 g/kg/day group (p = 0.019) across timepoints. The protein goal of 2.0 g/kg/day was achieved by 30% of the participants at week 6 and 35% at week 12. | - None reported related to the intervention | - Half of all participants (20/40) maintained or gained muscle mass by week 12. - Irrespective of group allocation, percent change in muscle mass from baseline trended toward a positive association with actual protein intake and suggested that an increase of 1.0 g/kg/day protein may result in a 1.6% increase in appendicular lean soft tissue index (p = 0.090). - An increase in protein consumption was associated with an increase in physical function as measured using the Short Physical Performance Battery (p = 0.014). |
Keum et al., 2021 [25] | Analyzed in a per-protocol analysis: 33 (17 intervention, 16 control) Sex: 20 males, 13 females Mean age: 61.5 years (range 34–78 years) Cancer type(s): pancreatic ductal adenocarcinoma | - 34/40 (85.0%) were retained post-intervention at 12 weeks; 33/40 had evaluable data. | - None reported related to the intervention | - All the study participants showed a significant improvement in nutritional status (PG-SGA), with no between-group differences; those who used the Noom app more showed a significant improvement in PG-SGA. - The “above average” app users showed an increase in body weight and BMI compared to the “below average” users. - There was no statistically significant difference in quality of life between groups as measured using EORTC; there was an improvement over time for the intervention vs. control group as measured using the global health status and quality of life scale (p = 0.004). - Skeletal muscle index decreased for the non-app user and increased for the “above average” app user (p = 0.041). - A decrease in tumor size was more prominent in the “above average” user vs. the non-users (p = 0.17). - There were no between-group differences in survival metrics. |
Lin et al., 2017 [26] | Analyzed N: 110 (55 intervention, 55 control) Sex: 70 males, 40 females Mean age: 52.6 years Cancer type(s): colorectal | - Retention not reported, implied 100% retention. | - None reported related to the intervention | - Body weight increased over time for the intervention group (p ≤ 0.001 at the 6th and 12th cycles) and not the control group (p > 0.24). - Nutritional biomarkers albumin and prealbumin increased over time for both groups, with higher increases in the intervention group. - There were no treatment interruptions from nutrition-related complications in the intervention group, but there were 8 cases in the control group. |
Loprinzi et al., 1996 [27] | Analyzed N: 107 (54 intervention, 53 control) Sex: all females Median age: 43 (range 26–57) years Cancer type(s): breast | - Data available for 107/109 (98.2%) who entered the study. | - None reported related to the intervention | - The median weight change for the intervention group was +2.0 kg and for control group it was +3.5 kg 6 months after start of chemotherapy (not statistically significant). - More weight gain was seen in participants with higher Quetelet’s indexes (p < 0.01), those who had been on a diet in the previous 6 months (p < 0.025), those with higher scores on an introversion scale (p < 0.04), and those with higher scores on an obsessive/compulsive scale (p < 0.025). - There was no evidence of association between any weight change variable and appetite, nausea, or vomiting. |
Najafi et al., 2019 [28] | Analyzed N: 137 (70 intervention, 75 control) Sex: all females Mean age: 46.9 ± 12.4 years intervention, 46.0 ± 8.8 years control Cancer type(s): breast | - Same as Abdollahi et al. [19] - The intervention group on average ate less fat and less protein over time and the control group did not, but comparisons to recommendations are not reported. | - None reported related to the intervention | - Quality of life was higher in the intervention vs. control groups and stable over time for both groups. - Nausea, as measured using three instruments, was less for the intervention group vs. the control group for all three chemotherapy sessions (p < 0.001). - Participants in the intervention group had higher levels of physical, role, emotional, and cognitive functioning across all three chemotherapy time points (p < 0.001 for all). - Participants in the intervention group had equal (dyspnea, insomnia, appetite loss, constipation, diarrhea) or less severity (fatigue, nausea and vomiting, pain) of all symptoms measured. |
Ollenschläger et al., 1992 [29] | Analyzed N: 29 (16 intervention, 16 control) Sex: 16 males, 16 females Age range: 17–59 years Cancer type(s): acute leukemia | - 29/32 (90.6%) analyzed. - The mean dietary intake of those in the intervention group was 23.3 ± 11.4 kcal/kg ideal body weight during weeks of weight loss, 30.9 ± 13.1 during weeks with stable weight, and 39.3 ± 12.2 kcal (p < 0.0001). - Dietary intake was not compared to goals for the intervention group and was not reported for the control group. | - It is mentioned that toxicities occurred, but none were stated to be attributed to the intervention. | - There were no between-group differences in septic episodes or days with body temperature > 38.5 °C. - Participants in both study groups lost body weight up to the third or seventh study week. Among the participants treated with LAM-6 (n = 13), those in the intervention group showed more weeks with weight gain vs. those in the control group. There were no between-group differences for participants undergoing other types of treatment. At the end of induction therapy, only 5/16 participants in the intervention group showed a body weight <95% of their pre-study weight vs. 11/16 control participants. - Neither within-group nor between-group changes were reported for well-being or quality of life. Weight loss was positively correlated with weakness/malaise, energy intake was negatively correlated with weakness/fatigue, energy intake was negatively correlated with side effects of therapy, and quality of diet was negatively correlated with side effects of therapy (all p < 0.01). |
Ovesen et al., 1993 [30] | Analyzed N: 117 (59 intervention, 58 control) Sex: 90 female, 27 male Mean age: 59 years (intervention), 58 years (control), range 22–80 years Cancer type(s): ovarian (n = 86), lung (n = 78), breast (n = 36) | - 105/137 (85.4%) analyzed. - Daily energy and protein intake did not change in the control group and increased significantly in the intervention group (~1 MJ [239 kcal] and 10 g of protein) at one month, which was sustained during the study period. - Dietary intake was close but did not meet goals for the intervention group, with a 1.5 ± 0.4 × basal energy expenditure and protein intake of 1.1 ± 0.4 g protein/kg vs. 1.3 ± 0.4 × basal energy expenditure and 0.9 ± 0.5 g protein/kg in the control group. | - No adverse events were reported. | - Both groups lost body weight in the first month of the study, but the intervention group gained more weight subsequently, but the difference did not meet statistical significance (p = 0.15). - In regard to tumor response, 65% were responders in the intervention vs. 69% in the control group at 3 months (p = 0.83). At 5 months, there were more responders in the intervention group (63%) vs. the control group (46%, p = 0.11). - There were no between-group effects on quality of life. |
Regueme et al., 2021 [31] | Analyzed N: 155 Sex: 78 males, 77 females Mean age: 77.3 ± 4.8 years Cancer type(s): colon (43), lymphoma (34), lung (11), ovary (16), breast (9), stomach (9), pancreas (17), prostate (8), bladder (6), cholangiocarcinoma (1), unknown (1) | - Same study as Bourdel-Marchasson et al. [21]. - 155/283 (54.8%) analyzed (57 died before end of chemotherapy, 71 had missing data). - Dietary intervention increased total energy and protein intake (specifics not reported) | - None reported related to the intervention | - There were no between-group differences in health-related quality of life; physical functioning and fatigue decreased with time in some participants (23.9% and 36.9%, respectively). - Worsening of comprehensive gerontological assessment items was observed frequently for the Geriatric Depression Scale (51.6% of participants), timed up-and-go (50.0%), OLS (76.5%), activities of daily living (14.9%), and instrumental activities of daily living (34.8%); there were no between-group differences. |
Sukaraphat et al., 2016 [32] | Analyzed N: 50 (25 intervention, 25 control) Sex: 34 male, 16 female Mean age: 61.3 years (intervention), 62.7 years (control) (range 45–81 years) Cancer type(s): lung (n = 35), cholangiocarcinoma (n = 15) | - 100% retention through post-intervention (12 weeks). - Energy intake was similar at baseline (mean 1415 kcal in the intervention group vs. 1510 in the control group, p = 0.48). Energy intake was greater in the intervention group than the control group at post-intervention (1832 ± 672 kcal vs. 1641 ± 343 kcal/day, p = 0.21) and 2 months post-intervention (1847 ± 443 kcal vs. 1615 ± 313 kcal/day, p = 0.06), but differences did not reach statistical significance. | - No adverse events were reported. | - The within-group change in body weight from pre- to post-chemotherapy was higher for the intervention group vs. the control group (2.3 ± 6.2% vs. −1.7 ± 6.2%, p = 0.03). Similar results were seen for BMI (difference in within-group changes p = 0.03). From pre-chemotherapy to 2 months post-chemotherapy, body weight increased 2.6 ± 6.4% in the intervention group and changed to −0.27 ± 7.1% in the control group (p = 0.19). - While scores were similar at baseline (p > 0.2), there was a lower PG-SGA score (better nutritional status) in the intervention vs. control group at post-intervention (p < 0.001) and 2 months post-intervention (p < 0.01). - Quality of life was similar at baseline and then greater in the intervention vs. control group at post-intervention (p = 0.01) but the significance did not hold 2 months post-intervention (p = 0.08). - There were no between-group differences in serum albumin or total lymphocyte count at baseline, post-intervention, or 2 months post-intervention. - There was a higher percentage of participants in the control group vs. the intervention group who delayed chemotherapy (48% vs. 36%), but the difference was not statistically significant (p = 0.57). - Response to treatment (stable/progression of disease, partial response) was similar between groups. - Infection rates were similar between groups (no statistics reported). |
Tang et al., 2024 (China) [58] | Analyzed N: 60 (30 intervention, 30 control) Sex: 45 males, 15 females Mean age: 53.8 years Cancer type(s): nasopharyngeal carcinoma | - 100% retention. - Effective recovery rate of 100% for 60 questionnaires. | - None reported related to the intervention | - NRS scores were higher for the control group than the intervention group throughout the 24-week study (week 3, p = 0.014; week 10, p = 0.070; week 24, difference 0.33 ± 0.77, p = 0.023), indicating higher nutritional risk. - Body weight of the control group was lower than that of the intervention group throughout the study (mean difference of 3.9 kg at 12 weeks, p = 0.044). - Quality of life (as measured by SF-36 Scale) after 24 weeks showed higher scores (higher quality of life) for the intervention group vs. the control group for physiological function, somatic pain, general health, energy, and social function (p < 0.05). |
van der Werf et al., 2020 [33] | Analyzed N: 72 (51 intervention, 54 control) Sex: 66 males, 41 females Mean age: 64 ± 13 years intervention, 66 ± 10 years control Cancer type(s): metastatic colorectal cancer | - 102/107 (95.3%) continued until T1 (after ~9 weeks of CAPOX(-B)/capecitabine(-B) or 12 weeks of FOLFOX(-B)). - 72/107 (67.2%) continued until T2 (mean 19 weeks). - The intervention group had a mean of 12.1 ± 4.6 dietetic consultations vs. 3.5 ± 2.5 in the control group. | - None reported related to the intervention | - 30% of the patients had a decrease in skeletal muscle area of ≥6.0 cm2 from T0 to T1, with no between-group differences in incidence (p = 0.47) or degree of change (p = 0.9 for both change from T0 to T1 and T1 to T2). - The intervention group increased in body weight from T0 to T1 1.7 kg (95% CI 0.0–3.3 kg) vs. −0.2 kg (−1.6–1.3 kg) in the control group. - There was no effect of group on muscle density, hand grip strength, quality of life global health score, physical functioning, or quality of life subscales. - There was no effect of group on the incidence of grade 3/4 toxicity or reduced treatment intensity. - Participants in the intervention group had significantly longer progression-free survival (p = 0.039) and overall survival (p = 0.046). |
Wang et al., 2023 [34] | Analyzed N: 36 (18 intervention, 18 control) Sex: 28 males, 8 females Median age: 67.5 (range 54–78) years Cancer type(s): esophageal cancer (squamous cell carcinoma, 34; adenocarcinoma, 2) | - 36/53 (67.9%) completed the study. | - None reported related to the intervention | - In the intervention group, albumin decreased at first then recovered and was similar to baseline (p = 0.21), though it decreased in the control group over time (p = 0.01); similar trends were observed for hemoglobin (p = 0.09 for the intervention and p = 0.04 for the control group over time). - C-reactive protein levels increased during treatment, but then gradually decreased in both groups, though increases were more in the control group compared to baseline (p = 0.02). - The incidence of grade ≥ II lymphocytopenia was higher in the control group than the intervention group (33.3% vs. 61.1%, p = 0.03). - The incidence of complications including nausea, vomiting, leukopenia, radiation esophagitis, and myelosuppression were similar between groups. - There was no between-group difference in remission status. - The average length of hospital stay was 12 days less (47 vs. 35 days, p = 0.001), and in-patient expenses were CNY 20,504 less (p = 0.004) in the intervention group compared to the control group. |
White et al., 2020 [35] | Analyzed N: 50 (24 intervention, 26 control) Completed the study: N = 40 (20 intervention, 20 control) Sex: 14 males, 36 females Mean age: 56.5 ± 13.9 intervention, 44.7 ± 14.4 control Cancer type(s): cervical (35), bladder (15) | - 100% remained at the study at 6 weeks and were therefore assessed for the primary outcome (treatment toxicity). - 48/50 (96%) had analyzable data at 12 weeks and 40/50 (80%) had analyzable data at 12 months. - It was possible to perform 57/72 (79%) of proposed intervention tests with no disruption of oncological management. - There was excellent compliance with questionnaires. - 151/179 (84%) of requested food diaries were completed. | - There were no adverse events attributable to the intervention. | - Participants in both groups experienced fewer gastrointestinal symptoms. At 6 weeks (primary endpoint), bowel scores were higher (worse) for the control vs. intervention group. - There were no grade 3–4 bowel toxicities in either group. - Chemotherapy interruptions due to bowel symptoms in 3 patients in the control group and 1 patient in the intervention group. - 17/24 (71%) participants in the intervention group received the planned chemotherapy vs. 16/26 (62%) in the control group. - There were no between-group differences in anxiety, depression, or quality of life. - Participant ratings of satisfaction were “very satisfied” or “extremely satisfied” in both groups. |
Xie et al., 2017 [36] | Analyzed N: 144 Sex: 85 males, 59 females Mean age: 55.5 ± 11.4 years control, 55.3 ± 8.6 years intervention Cancer type(s): gastric cancer (stomach, 74; cardia, 31; antrum, 39). | - All randomized participants were included in analyses (100%). - The intervention group had a significantly higher calorie intake, ratio to recommended nutritional intake (RNI), and iron intake vs. the control group (all p < 0.05 for all) at the first chemotherapy session. | - None reported related to the intervention | - Hemoglobin was similar between groups at baseline and was higher in the intervention vs. control group for the 3rd–8th chemotherapy session (p < 0.05 for all time points). - Serum total protein was higher at the first chemotherapy session for the intervention vs. control group (p = 0.021) but similar for all other time points. - Albumin tended to be higher for the intervention vs. control and reached statistical significance at some time points. - Reductions in body weight were slightly attenuated in the intervention vs. control group, but differences did not reach statistical significance. - Chemotherapy interference was lower in the intervention group than the control group (completion of scheduled regimen: 73.61% vs. 55.56%, p = 0.024). |
Nutrition counseling plus exercise | ||||
Allen et al., 2022 [37] | Analyzed N: 48 (26 intervention, 28 control) Sex: 46 males, 8 females Mean age: 64 ± 8 years Cancer type(s): advanced esophagogastric cancer (adenocarcinoma, 48; squamous cell carcinoma, 6) | - 48/54 (92%) completed the study. - Attendance at the supervised exercise sessions was 76 ± 14% and adherence to the home exercises was 65 ± 27%; medical coaching adherence was 82 ± 20%. | - No intervention-related adverse events were reported to study staff. | - There was no between-group difference in anaerobic threshold (p = 0.57) or group×time interaction (p = 0.40). - Reductions in peak VO2 were attenuated in the intervention group following neoadjuvant therapy (p = 0.022). - The intervention group had less skeletal muscle loss following neoadjuvant therapy (p = 0.049). There was no between-group difference in the incidence of sarcopenia (p = 0.404). - Decreases in hand grip strength were attenuated in the intervention vs. control group (p = 0.009). - Global health status quality of life was higher for the intervention group vs. the control group (p = 0.002 for group × time). - Anxiety and depression were lower in the intervention vs. control group preoperatively and postoperatively at 6 weeks and 6 months. - More intervention participants completed chemotherapy at the full dose (75% vs. 46%, p = 0.036). - Pulmonary complications occurred at a similar rate between groups (59% intervention vs. 78% controls, p = 0.079). - There were no between-group differences in median length of hospital stay, 30-day readmission, or 3-year postoperative cancer-related mortality. |
Basen-Engquist et al., 2020 [38] | Analyzed N: 37 (19 intervention, 18 control) at the end of the study Sex: all female Mean age: 49 years Cancer type(s): breast | - 73.7% retention (28/38). - 12/19 (63%) controls and 16/19 (84%) completed the assessment at T2 (end of chemotherapy). - Intervention participants attended an average of 80% of the in-person sessions and 43% of the telephone sessions. - 88% (n = 8) of participants reported at T2 that they were satisfied with the intervention, and 88% (n = 8) said they would recommend it to a friend. | - None reported related to the intervention | - Body weight fluctuated over the course of the study, and there was no effect of group (p = 0.083). - Waist circumference was significantly lower across the study in the intervention group compared to control (p = 0.019). - In regard to quality of life, the intervention group reported significantly greater improvements on the SF-36 vitality subscale than control participants at the end of chemotherapy (p = 0.005); there were no significant intervention effects for physical functioning, mental health, role physical, bodily pain, general health, social functioning, role emotional, or body image (p > 0.1). |
Brouwer et al., 2024 (Netherlands) [59] | Analyzed N: 81 (40 intervention, 41 control) Sex: all female Average age: 59 years Cancer type(s): ovarian | - Feasibility described in Kenkhuis et al. [60]. | - Safety reported in Kenkhuis et al. [60] | - The proportion of patients achieving a relative dose intensity ≥ 85% was 74.4% in the intervention group compared with 61.5% in the control group (odds ratio 2.04, 95% confidence interval 0.75 to 5.84). - The intervention effect on progression free survival was not statistically significant (hazard ratio 1.63, 95% confidence interval 0.82 to 3.23). At 18 months, the proportion of patients without disease progression was 73% in the intervention group and 51% in the control group (no statistics presented). |
Carayol et al., 2019 [39] | Analyzed N: 135 Sex: All female Mean age: 52 years Cancer type(s): breast | - Overall retention was 135/143 (94.4%). - 72/72 (100%) of the intervention group and 63/71 (89%) completed the intervention period (26 weeks) and provided patient-reported outcomes. - Adherence to the Adapted Physical Activity and Diet (APAD) intervention was 67% for completed planned exercise sessions and 97% for completed scheduled diet sessions. | - No serious adverse events were attributed to exercise or diet intervention. | - Fatigue peaked during treatment for both groups. All dimensions of fatigue were significantly less for the intervention vs. control group at the end of chemotherapy and the end of radiotherapy (end of the intervention, except for general fatigue, p < 0.03). Fatigue was similar between groups at the 6-month follow-up but was higher in the control vs. intervention group at 1 year (p < 0.05). - Quality of life was lowest during treatment for both groups. Those in the intervention group had higher quality of life at all time points after baseline (p < 0.05). - Anxiety and depression were less for the intervention group vs. control at the mid- and post-intervention times points, but not at the 6-month or 1-year follow-up. - In regard to muscle function, lower-limb muscle endurance was higher in the intervention vs. control group at the end of radiotherapy; there were no between-group differences in power or force. - There were larger, favorable decreases in body weight and body mass index in the intervention vs. control group at the end of radiotherapy, with no differences in total muscle mass. - Chemotherapy completion rates were similar between groups (65.1% in the control group and 78.8% in the intervention group, p = 0.083). |
Demark- Wahnefried et al., 2008 [40] | Analyzed N: 82 (27 calcium-rich (control), 26 calcium-rich plus exercise, 29 calcium-rich plus high fruits and vegetables, low fat + exercise) Sex: all female Mean age: 41.8 ± 5.6 Cancer type(s): breast | - 82/90 (91.1%) completed the study. - Attendance at the telephone counseling sessions was control > diet + exercise > exercise (p = 0.071). - More logs were submitted by the control group vs. the exercise and diet + exercise groups. - Calcium intake increased in all arms of the study (p = 0.02, baseline vs. 3 and 6 months); it was similar among all groups with 74–87% consuming ≥1200 mg per day. - The diet + exercise group had higher fruit and vegetable intakes and lower fat intakes at 3 months and 6 months vs. baseline; 16% in the diet + exercise group consumed ≤30% energy from fat and 39% reported intakes of ≥5 servings of fruit and vegetables per day. - The control diet received the highest feasibility scores, ranging from 1.3 to 1.5 (“very likely” to “likely”). | - No adverse events related to the intervention were reported. | - All measures of adiposity increased over time among all groups, with the exception of waist circumference, which decreased in the diet + exercise arm; at 6 months, the diet + exercise arm also had lower scores for percentage of body fat. - There were no within-group changes over time or between-group differences in lean body mass. - In regard to bone density, spine and hip t-scores were slightly lower at 6 months than at baseline for all arms, with no between-group differences. - In regard to serologic biomarkers (insulin, proinsulin, insulin-like growth factor-1, C-reactive protein, total cholesterol, high density lipoprotein cholesterol, low density lipoprotein cholesterol, sex hormone binding globulin, interleukin-1β, tumor necrosis factor-α receptor 2), there were no between-group differences. - Significant improvements in quality of life were observed over time among all groups, with no between-group differences. - No within-group differences over time or between-group differences were observed for depression or anxiety. |
Djuric et al., 2011 [41] | Analyzed N: 30 Sex: all females Mean age: 52 Cancer type(s): breast | - 30/40 (75%) completed the study. - 70% of self-monitoring logs were submitted, only 2 participants submitted < 50% of requested logs. - The number of servings of fruits and vegetables per day increased in the intervention arm, and the mean reported intake at 12 months was just above the minimum intervention goal of 7 servings/day. - Serum total carotenoids increased in the intervention arm. - In the intervention arm, fruit and vegetable intakes at 12 months were 9.8 servings/day by screener vs. 7.3 servings/day by recalls, confirming over-estimates by the screener. | - None reported related to the intervention | - Anthropometrics: body weight, body fat, and waist circumference tended to decrease in the intervention group and increase in the control group. - Well-being improved in both study arms and the change reached statistical significance only in the intervention arm. - Small, favorable improvements in insulin, glucose, and leptin were noted in the intervention vs. control arm. - Participants in the intervention arm declared that the study was helpful, while those in the control arm said helpfulness was neutral. |
Jacot et al., 2020 [42] | Analyzed N: 180 in the intent-to-treat analysis, 179 in the per-protocol analysis Sex: all female Mean age: 52.5 ± 9.9 years Cancer type(s): breast | - 89.2% analyzed. - 67/142 (47.2%) were at least 80% adherent. - The intervention increased physical activity (metabolic equivalents per week, p < 0.001) and engagement in moderate intensity recreational activities (p < 0.001) - Participants in the intervention vs. control arm more frequently met WHO dietary recommendations at T1 (end of chemotherapy, 81.4% vs. 61.9%, p < 0.001), 80.5% vs. 70.9%, p = 0.071), and T3 (86.6% vs. 68.1%, p = 0.002). | - None reported related to the intervention | - Compared to T0, general fatigue was similar between groups at T1 (end of chemotherapy, p = 0.274), T2 (end of radiation therapy, p = 0.157), and T3 (1 year after enrollment, p = 0.933). Physical and mental fatigue (from the Multidimensional Fatigue Inventory) and the symptom fatigue increased over time in both arms, but there were no between-group differences. In the per-protocol analysis, the intervention enhanced motivation (p = 0.003) and reduced mental fatigue (p = 0.069). - There were no between-group differences in quality of life. - Emotional function increased and cognitive function decreased over time. - A lower proportion of patients in the intervention arm (54.0%) vs. control arm (66.9%) had depression 1 year after enrollment. - There were no between-group differences in body weight. |
Kenkhuis et al., 2024 (Netherlands) [60] | Analyzed N: 81 (40 intervention, 41 control) Sex: all female Average age: 59 years Cancer type(s): ovarian | - 64/81 (79%) retention. - In the intervention group, median attendance for the physiotherapist was 71.7%. - Among the 39 participants with available physical activity logs, 16 participants attended more than 75% of the prescribed exercise sessions. - The median exercise relative dose intensity ranged from 0 to 97.3% (mean 72.6%, SD 16.0). - Median attendance for dietary sessions was 80% (IQR: 50–100%), and among the 31 participants with data, 18 participants attended more than 75% of the prescribed dietary sessions. - There were no within-group or between-group differences in adherence to the WCRF/AICR lifestyle recommendations, the Dutch Healthy Diet Index, caloric intake, or protein intake. | - No adverse advents were recorded during the study. | - There were within-group increases in fat-free mass and physical functioning and decreases in fatigue over time, with no between-group differences. - No significant difference between the groups were found for FFM (β = −0.5 kg; 95% CI = −3.2; 2.1), physical functioning (β = 1.4; 95% CI = −5.4; 8.3), or fatigue (β = 0.7; 95% CI = −1.5; 2.8). - No between-group differences were found for physical fitness, health-related quality of life, anxiety, depression, neuropathy, or sleep disturbances. |
Maurer et al., 2022 [43] | Analyzed N: 11 (5 intervention, 6 control) Sex: all females Median age: 58 (range 21–77 years) Cancer type(s): ovarian | - 11/15 (73.3%) participants completed the study. - Adherence to the nutrition intervention was 76.8% (phase I, 92.3%; phase II, 59.6%). - The intervention group increased their protein intake from 65.8 g/day at baseline to 107.9 g/day at the end of chemotherapy; this increase was greater than for the controls. - The intervention group increased calorie intake from 1860 kcal/day at baseline to 2389 kcal/day at the end of chemotherapy. - The intervention group increased the Mediterranean diet adherence score from a median of 7 at baseline to a median of 10 at week 52; scores were similar between groups. | No adverse events reported to the study team due to the intervention or in-person assessments. | - Participants in the intervention group increased their hand grip strength from a median of 22.0 kg to 24.8 kg, while the control group only increased from 21.8 kg to 22.4 kg. - Health-related quality of life increased from baseline to 52 weeks from 37.5 to 70.8 points in the intervention group and from 41.7 to 50.0 score points in the control group. - Both total and physical fatigue decreased from baseline to 52 weeks in both groups, and the decrease was larger in the intervention group. |
Puklin et al., 2024 (United States) [61] | Analyzed N: 173 (87 intervention, 86 control) Sex: all female Mean Age: 53 ± 11 years Cancer type(s): breast | - A total of 149 women (86%) completed the 1-year (80 [92%] intervention; 69 [80%] control). - Among the 150 women with dietary assessments at baseline, 128 (85%) completed them at 1 year (72 [89%] intervention; 56 [81%] control). - On average, women in the intervention arm attended 15 (SD = 3) of 16 counseling sessions during the 1-year intervention. - Ninety percent of women attended at least 80% of the sessions (≥13 sessions) and 89% attended all 16 sessions. | - No adverse events were attributable to the intervention. | - At 1 year, 76% of women randomized to intervention adhered to the physical activity guideline compared with 24% of control women (p < 0.001). Over the year-long intervention, the intervention arm increased 136.1 min/week (p < 0.001) of physical activity more than the control arm. - Over the year-long intervention, those in the intervention group increased diet quality a mean of 4.9 points vs. 2.4 points in the control group (p = 0.08, not significant). In regard to individual dietary components, the intervention group consumed less added sugar than the control group (p = 0.02), though there were no other statistically significant differences. - Among women in the intervention arm, in the multivariable model for change in minutes/week of physical activity over 1 year, lower baseline fatigue was associated with greater improvement in minutes/week of physical activity (p = 0.04). |
Raghunath et al., 2020 [44] | Analyzed N: 116 (58 intervention, 58 control) Sex: 58 males, 58 females Median age: 48 (range 18–65) years Cancer type(s): colorectal cancer | - 118/120 (98.3%) completed the study; two in the control arm developed recurrences and were removed from the analysis; two from the intervention group were also removed so the groups would be equal. | - None reported related to the intervention. | - Hematological parameters maintained similar patterns between groups over the course of treatment and post-treatment survivorship. - The intervention vs. control group exhibited lower levels of the tumor marker carcinoembryonic antigen after chemotherapy (p = 0.0003). - Symptoms of anxiety, depression, and distress, and symptom severity, increased for the control group and decreased for the intervention group over the course of the study (all p < 0.001). The Functional Living Index: Cancer (FLIC) decreased for the control group and increased for the intervention group (p < 0.0001). |
Sanft et al., 2023 [45] | Analyzed N: 173 (87 intervention, 86 control) Sex: all female Mean age: 53 ± 11 years Cancer type(s): breast | - 173/173 (100%) analyzed. - There was 91% attendance at counseling sessions. - The intervention group increased servings of fruit and vegetables and dietary fiber, while the usual care group decreased intake over the course of chemotherapy/the study (p < 0.01, between-group difference). | - 55% in the intervention group and 63% in the control group experienced at least one nutrition impact symptom (p = 0.32), attributed to chemotherapy. - No adverse events were attributable to the intervention. | - Both groups experienced decreases in body weight (average 1 kg, p = 0.76 between groups). - Relative dose intensity was 92.9 ± 12.1% in the intervention group and 93.6 ± 11.1% in the control group (p = 0.69). - Among the 72 women who received neoadjuvant (vs. adjuvant) chemotherapy, those in the intervention arm were more likely to have a partial complete response (53% vs. 28%, p = 0.037). |
Stelten et al., 2022 [46] | Analyzed N: 24 Sex: all females Mean age: 53.7 ± 10.6 years Cancer type(s): ovarian | - 20/24 (83.3%) completed the study. - Interviews were available for 14 participants in the intervention group and 10 in the control group. - Participants attended 33–133% (median 100%) of the prescribed dietary sessions. | - None reported related to the intervention. | - Patients appreciated guidance on exercise and nutrition and perceived benefits including improved physical fitness, quality of life, peer support, and recovery after surgery and/or chemotherapy cycles. |
Zhang et al., 2023 [55] | Analyzed N: 97 (51 intervention, 46 control) Sex: 72 male, 26 female Mean age: 61.6 ± 9.6 years (intervention), 62.4 ± 8.1 (control) Cancer type(s): squamous cell carcinoma (n = 28), adenocarcinoma (n = 50), small cell or other lung cancer (n = 20) | - 102/106 (96.2%) were retained at post-intervention/post- chemotherapy (12 weeks). - Energy intake was similar between groups. Protein (g/day), fat (g/day), fiber, omega-3 fatty acids, and many micronutrients were significantly greater in the intervention vs. control groups (p < 0.001). Carbohydrates and cholesterol intakes were greater in the control vs. intervention groups (p < 0.001). | - No adverse events were reported. | - Cancer-related fatigue did not differ at baseline, week 4, or week 8 (all p > 0.05) but was significantly lower at week 12 (p < 0.01); fatigue decreased over time for both groups (p < 0.01) and there was a group×time interaction favoring the intervention group (p < 0.001). - For high-sensitivity C-reactive protein, a marker of inflammation, levels decreased over time for both groups from baseline through week 6 and week 8, then increased at week 12 for the control group only (p < 0.001 at week 12). The group×time interaction was significant (p < 0.001), favoring the intervention group. - Nutritional status tended to increase over time as measured with PG-SGA (p < 0.001 for time). The intervention group had lower PG-SGA levels (better nutritional status) at week 8 (p = 0.007) and week 12 (p = 0.03) but there was no group×time interaction. - Serum albumin levels tended to increase with time for the intervention group and remain stable for the control group (group × time interaction p = 0.048). - The intervention was associated with greater quality of life, especially emotional functioning, cognitive functioning, fatigue, and sleep disturbance (between-group differences p < 0.05 at week 12). |
Nutrient-specific dietary patterns | ||||
Bille et al., 2018 [47] | Analyzed N: 32 (with weight measurements) Sex: 18 males, 14 females Mean age: 52 ± 14 (with full registration of intake) 52.5 ± 16 (without full registration of intake) Cancer type(s): Acute and chronic leukemia and lymphoma, Hodgkin’s lymphoma, and non-Hodgkin’s lymphoma (specifics not described) | - 32/62 (51.6%) who signed the consent completed the study. - 18 patients had full registration of their intake and 14 patients had incomplete registrations. - All 32 participants who started the study completed it. - Compliance from weeks 1 to 4 (details not presented). | - None reported related to the intervention. | - Participants gained body weight (1.2 ± 1.9 kg, 2%) during the intervention period and lost weight (−2.8 ± 5.2 kg, 4%) in the control period. - Chicken in curry was the dish most frequently consumed in the home-cooked meals, and curry soup was the most frequently consumed prepared dish. |
Harvey et al., 2023 [6] | Analyzed N: 29 (21 intervention, 8 control) Sex: 1 male, 28 female Mean age: 51.0 ± 15.1 years Cancer type(s): breast (27), other (2) | - Feasibility described in Kleckner et al. [52]. | - Safety reported in Kleckner et al. [52]. | - Facilitators to adherence included education, convenience, and empowerment. - Barriers to adherence included chemotherapy side effects, food preferences, and lifestyle changes. |
Gardner et al., 2008 [48] | Analyzed N: 153 (78 intervention, 75 control) Sex: not reported Mean age: 64 years (range 17–88 years, intervention), 63 years (range 47–84 years, control) Cancer type(s): Acute myeloid leukemia (144), myelodysplastic syndrome (9) | - 100% of participants completed the study. - Compliance was noted with daily food diaries. All patients remained on the correct diet, although some in the control group did not eat a fresh fruit or vegetable every day. | - None reported related to the intervention. | - 29% in the intervention (cooked) group and 35% of patients in the control (raw) group developed a major infection (p = 0.60), and the rate was similar to incidence rates for people not randomized in the study. - Complete response rates were similar between groups (56% in the intervention, 61% in the control, 64% in the non-randomized). - 53% in the intervention and 57% control had an antecedent hematologic disorder. - The number of days spent with neutrophil counts < 500/µL and <100 µL were similar between groups (p = 0.77 and p = 0.53, respectively). - 28 patients in both the cooked and raw groups lost weight (a median of 5 lb in each group) over the course of the study. - Incidence of grade 3 or 4 mucositis or diarrhea was similar between groups. - Fever of unknown origin occurred in 51% of the cooked group and 36% of the raw group. |
IJmker-Hemink et al., 2021 [49] | Analyzed N: 13 (7 control, 9 intervention) Sex: 6 males, 14 females at baseline Mean age: 59 ± 14 years control, 63 ± 10 years intervention Cancer type(s): gastrointestinal (8), breast (4), gynecological (8) | - 13/20 (65%) completed the study after 35% dropouts. - 90% of participants completed their symptom diaries. | - None reported related to the intervention. | - Of 17 participants, 88% (n = 15) graded the level of burden (from 0 to 10) that they experienced by participating in the pilot study; the median level of burden was 2, ranging from 0 to 3. - The main reason for participating was to help future patients. |
IJmker-Hemink et al., 2023 [50] | Analyzed N: 116 (69 control, 67 intervention) Sex: 38 male, 98 female Mean age: 59.0 ± 13.9 years control, 62.9 ± 9.5 years intervention Cancer type(s): breast (40), colorectal (39), gynecological (28), other (19) | - 136/148 (92%) completed randomization, 116/148 (78.3%) completed the intervention, and 96/148 (64.9%) completed the 3-month follow-up assessments. - Energy intake was 21% higher in the intervention group after 3 weeks of the intervention (p < 0.05). In the intervention group, 31% met protein requirements before the intervention and 47% met them after, compared to 28% and 33% of controls at the same time points. | - None reported related to the intervention. | - After the intervention, quality of life was 5.0 points higher for the intervention vs. control group (p = 0.122); the score was similar between groups at follow-up. - There were no within-group or between-group differences in handgrip strength or nutritional status (PG-SGA). - The Short Physical Performance battery increased in the intervention group from pre- to post-intervention (p < 0.05). - Emotional functioning was higher in the intervention vs. control group after the intervention (p < 0.05). Pain and tiredness were lower in the intervention vs. control group after the intervention. There were no other between-group or within-group differences in functional or symptom scales. |
Jalali et al., 2018 [51] | Analyzed N: 50 (25 intervention, 25 control) Sex: 29 males, 21 females Mean age: 41.2 ± 14.2 years intervention, 40.8 ± 12.7 years control Cancer type(s): acute myeloid leukemia | - No dropouts reported, implying 100% retention. | - None reported related to the intervention. | - Serum albumin increased significantly more in the intervention group vs. the control group over the course of the study (p = 0.01); albumin decreased with time in the control group (p = 0.03). - Protein intake increased significantly in the intervention group from pre- to post-study (p = 0.001), but not in the control group (p = 0.2), though intake was similar between groups at the post-study time point (p = 0.26). - Body weight increased in the intervention group over time (p = 0.049) and decreased slightly in the control group (p = 0.13); weight was similar post-intervention. |
Kleckner et al., 2022 [52] | Analyzed N: 33 (23 intervention, 10 control) Sex: 2 males, 31 females Mean age: 51.0 ± 14.6 y Cancer type(s): breast (n = 30), other (3) | - 100% of the participants completed the study. At week 4, diet adherence scores increased from baseline by 2.7 ± 3.2 points for the intervention group and 0.7 ± 2.4 points for the control group. | - None reported related to the intervention. | - The intervention had small–moderate beneficial effect on cancer-related fatigue at week 4 (effect size = 0.31; 95% CI = −0.44–1.06) and week 8 (effect size = 0.25; 95% CI = −0.50−0.99). - There were small between-group differences in quality of life, favoring the intervention group (effect size = 0.26; 95% CI = 0.55−0.94). - There were no within-group changes or between-group differences in body weight, lipid profiles, homocysteine, or fructosamine. - There were no between-group differences in mitochondrial function (basal respiration, maximal respiration, or spare capacity). |
Sathiaraj et al., 2023 [53] | Analyzed N: 103 (52 intervention, 51 control) Sex: all females Mean age: 52 ± 9.7 y Cancer type(s): breast | - Retention not stated, implied to be 100%. - 50/52 (96%) of those in the intervention group adhered to the personalized nutritional counseling and 47/52 (90%) adhered to the whey protein supplement intake. | - None reported related to the intervention. | - Fatigue decreased over time in the intervention group and increased in the control group (p < 0.001). - The intervention group reported greater reductions in total energy and increases in protein and fiber intake (p < 0.001 for all three at post-intervention). - Body mass index decreased by 0.7 ± 0.8 kg/m2 in the intervention group, and the decrease was 0.4 ± 1.3 kg/m2 in the control group (p = 0.015). - Fat mass decreased in the intervention group (p < 0.001), with no within-group change in the control group. - Muscle mass decreased in the control group and was maintained in the intervention group (p < 0.05). Handgrip strength decreased slightly over time in the control group and was maintained in the intervention group, but differences were not statistically significant (p = 0.238). - Chemotherapy completion rate: relative dose intensity was similar between groups (91% having >90% in both groups). |
Villarini et al., 2012 [54] | Analyzed N: 94 (47 intervention, 47 control) Sex: all females Mean age: 52.7 ± 10.8 years intervention, 48.4 ± 9.4 years control Cancer type(s): breast | - 94/96 (97.9%) completed the study. - As for the 24 h recalls collected between chemotherapy administration, the women in the intervention group showed a significantly higher frequency of the consumption of whole grain cereals (2.2 vs. 0.8 times per day) and legumes (0.5 vs. 0.3 times per day), and a significantly lower frequency of the consumption of sugar (0.4 vs. 0.9 times per day), refined cereal products (1.1 vs. 2.1 times per day), and dairy products (0.5 vs. 0.9 times per day) compared with the control group (p < 0.01 for all). | - None reported related to the intervention. | - The intervention group showed a significant reduction in body weight (2.9 kg on average) vs. 0.1 kg in the control group (p = 0.0001 for difference in pre-post changes). - Women in the intervention group lost 2.29 kg of fat mass vs. 0.69 kg in the control group. (p = 0.03). Women in the intervention group lost 0.67 kg fat-free mass vs. 0.06 kg in the control group (p = 0.01). - In regard to hematological parameters, red blood cells, white blood cells, platelets, and hemoglobin levels significantly decreased between baseline and the end of treatment in both intervention and control groups, with no between-group differences. |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2024 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
James, S.; Oppermann, A.; Schotz, K.M.; Minotti, M.M.; Rao, G.G.; Kleckner, I.R.; Baguley, B.J.; Kleckner, A.S. Nutritional Counseling During Chemotherapy Treatment: A Systematic Review of Feasibility, Safety, and Efficacy. Curr. Oncol. 2025, 32, 3. https://doi.org/10.3390/curroncol32010003
James S, Oppermann A, Schotz KM, Minotti MM, Rao GG, Kleckner IR, Baguley BJ, Kleckner AS. Nutritional Counseling During Chemotherapy Treatment: A Systematic Review of Feasibility, Safety, and Efficacy. Current Oncology. 2025; 32(1):3. https://doi.org/10.3390/curroncol32010003
Chicago/Turabian StyleJames, Shalet, Alexie Oppermann, Kaitlin M. Schotz, Mackenzie M. Minotti, Gautam G. Rao, Ian R. Kleckner, Brenton J. Baguley, and Amber S. Kleckner. 2025. "Nutritional Counseling During Chemotherapy Treatment: A Systematic Review of Feasibility, Safety, and Efficacy" Current Oncology 32, no. 1: 3. https://doi.org/10.3390/curroncol32010003
APA StyleJames, S., Oppermann, A., Schotz, K. M., Minotti, M. M., Rao, G. G., Kleckner, I. R., Baguley, B. J., & Kleckner, A. S. (2025). Nutritional Counseling During Chemotherapy Treatment: A Systematic Review of Feasibility, Safety, and Efficacy. Current Oncology, 32(1), 3. https://doi.org/10.3390/curroncol32010003