Skip to main content
Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
J Cachexia Sarcopenia Muscle. 2022 Feb; 13(1): 55–67.
Published online 2022 Jan 10. doi: 10.1002/jcsm.12900
PMCID: PMC8818640
PMID: 35014216

Physical exercise at the crossroad between muscle wasting and the immune system: implications for lung cancer cachexia

Abstract

Cachexia is a syndrome characterized by involuntary weight loss and wasting of skeletal muscle mass. It is associated with worse overall survival and quality of life. The cancer‐induced systemic inflammation and the consequent host derived catabolic stimuli, trigger cachexia by inhibiting muscle protein synthesis and enhancing muscle catabolism. The muscle itself may further promote chronic inflammation, introducing a vicious catabolic circle. Nutritional support alone plays a limited role in the treatment of cancer cachexia and should be combined with other interventions. Physical exercise lowers systemic inflammation and promotes muscle anabolism. It also attenuates the age‐related physical decline in elderly and it might counteract the muscle wasting induced by the cancer cachexia syndrome. This review describes how cancer‐induced systemic inflammation promotes muscle wasting and whether physical exercise may represent a suitable treatment for cancer‐induced cachexia, particularly in patients with non‐small cell lung cancer. We summarized pre‐clinical and clinical studies investigating whether physical exercise would improve muscle performance and whether this improvement would translate in a clinically meaningful benefit for patients with cancer, in terms of survival and quality of life. Moreover, this review describes the results of studies investigating the interplay between physical exercise and the immune system, including the role of the intestinal microbiota.

Keywords: Cachexia, Systemic inflammation, Physical exercise, Immune system, Gut microbiota

Introduction

Cachexia is a metabolic disorder characterized by anorexia, involuntary weight loss, wasting of skeletal muscle mass and decreased muscle strength. 1 Cachexia is associated with worse overall survival (OS) and treatment tolerability; it increases fatigue, depression and decreases quality of life (QoL). 2 Cachexia typically associates with chronic systemic inflammation in chronic diseases, such as chronic obstructive pulmonary disease (COPD), 3 rheumatoid arthritis, 4 chronic kidney disease, 5 and especially cancer. 6 Patients with cachexia often present with elevated serum inflammatory markers, such as C‐reactive protein (CRP) and interleukin‐6 (IL‐6). 1 The muscle wasting occurring in patients affected by cachexia is called sarcopenia. 1 Sarcopenia is a consequence of the systemic inflammation that characterizes the cachexia syndrome. Systemic inflammation has shown to lead to muscle wasting and fat depletion. The cancer‐induced systemic inflammation and the consequent host‐derived catabolic stimuli trigger cachexia by inhibiting muscle protein synthesis and enhancing muscle catabolism (Figure 1 ). 10 The muscle itself exerts paracrine and endocrine effects by secreting myokines 11 that may further promote chronic inflammation and muscle wasting, 9 thus introducing a vicious catabolic circle. In patients with sepsis, systemic inflammation increases the catabolic processes already in the early phases of the infection. 12 Patients with metastatic cancer—and consequent more pronounced cancer related inflammation—have a higher risk of developing cancer cachexia. However, cachexia occurs already in patients with stage I non‐small lung cancer (NSCLC), even without any impairment in caloric intake. 13 About 20% of patients with early stage NSCLC present with signs of cachexia. 14 In patients with stage III NSCLC undergoing curative treatment, weight loss was observed already during the first 3 weeks of concurrent chemo‐radiotherapy (CCRT), before any caloric intake decrease was reported. 15 Adequate nutrition and caloric intake are helpful in avoiding body‐weight loss, but nutritional support alone has failed to counteract cachexia in several trials, consistently with the inability of nutrition to revert the catabolic process initiated by cancer‐induced inflammation. 16 Cachexia differs from fasting in which the organism proportionally preserves muscle integrity, favouring the use of adipose tissue for providing energy. 17 All together, these findings highlight that the key factor determining cachexia—and consequent weight loss—is systemic inflammation and not caloric intake. Therefore, nutrition serves as necessary materials for muscle build‐up, but it cannot reverse muscle wasting alone. Physical exercise lowers systemic inflammation (Figure 1 ) and promotes muscle anabolism. 18 It also attenuates the age‐related physical decline in elderly and therefore it might counteract the muscle wasting induced by the cancer cachexia syndrome. 19 Herein we describe how cancer‐induced systemic inflammation leads to muscle wasting and whether physical exercise in lung cancer may represent a suitable treatment for cancer‐induced cachexia. In the present work, we explore the link between cachexia and the immune system, pointing out why physical exercise could represent a crosslink between cachexia and the immune system, counteracting chronic inflammation. To the best of our knowledge, no extensive work has analysed the mutual relationship between the immune system, cancer related cachexia and physical exercise. We focus specifically on lung cancer, as in lung cancer, cachexia represent an issue across all the disease stages and deeply affects the prognosis. 20 Moreover, the majority of the data regarding the interplay between the immune system and cachexia have been studied in patients with lung cancer. We also present data investigating cachexia in other cancer types or other diseases, to illustrate mechanistically the cachexia pathways, and explain how physical exercise could be of benefit.

An external file that holds a picture, illustration, etc.
Object name is JCSM-13-55-g001.jpg

The interplay between muscle wasting, immune system and physical exercise cancer‐related chronic inflammation enhances the production of TNF‐α, IL‐6, IL‐1 and IL‐2. TNF‐α and IL‐1 activate NF‐κB, which impairs muscle build‐up. 7 IL‐6 directly promotes muscle wasting when TNF‐α co‐stimulation is present. 8 In response to exercise, muscle cells secrete IL‐6—in a pulsatile manner—IL‐7, IL‐10, and IL‐15. 9 Consequently, physical exercise increases CD8 + T cells, natural killer (NK) T cells and promotes the mobilization of B cells. Secretion of IL‐10 promotes macrophage M2‐polarization, which can be helpful for the muscle build‐up process. 10 The muscle breakdown process itself lowers the levels of circulating IL‐7 and IL‐15, promoting chronic inflammation. Chronic inflammation results in lower levels of circulating CD8 + T cells and NK T cells. 9 The runner symbolizes physical activity in general, not only endurance training.

Aetiology of cancer cachexia

Inflammatory cytokines (TNF‐α, interleukin [IL]‐6, and IL‐1), produced by the tumour and by the host in response to the tumour, lead to muscle wasting. The activation of Nuclear Factor kappa‐light‐chain‐enhancer of B cells (NF‐κB) might also antagonize the muscle synthesis process. 7 IL‐6 can enhance the acute immune response against the tumour, recruiting CD8 + T cells and clearing T regulatory (Treg) cells, when secreted in a pulsatile manner. However, it promotes muscle wasting when TNF‐α co‐stimulation is present, like in cancer‐induced inflammation. In addition to cancer itself, several and potentially coexisting pathophysiological mechanisms contribute in promoting inflammation and cachexia (Figure 2 ). 10 Anorexia—loss of appetite—occurs in about 30% of patients with lung cancer who receive a platinum doublet chemotherapy. 21 It impairs food intake—therefore limiting the resources for muscle build‐up—and might increase circulating IL‐6 and TNF‐α, therefore enhancing systemic inflammation, as shown in patients with anorexia nervosa. 22 In patients with cancer, it is particularly challenging to determine the aetiology of cancer cachexia since anti‐cancer treatment itself can enhance systemic inflammation and promote muscle wasting. 23 Chemotherapy directly causes skeletal muscle depletion by activating the NF‐κB pathway 23 and by enhancing the production of pro‐inflammatory cytokines (IL‐1β, IL‐6, and TNF‐α). 15 Radiation therapy contributes to muscle wasting through off‐target radiation to muscle fibres. 24 Although radiation impairs muscle regenerative capacity and promotes fibrosis locally, it is unlikely that systemic muscle wasting evolves as a direct effect of local irradiation. The systemic effects are likely mediated by an inflammatory reaction triggered by radiation. In mouse models, radiation therapy increases the intramuscular production of IL‐6, enhancing chronic inflammation, which may be modifiable through physical exercise. 25 This represents an additional incentive to consider physical exercise to modulate muscle wasting in patients with lung cancer. Decreased physical activity, which is typical of patients with cancer, could also translate into deconditioning and muscle mass depletion. 26

An external file that holds a picture, illustration, etc.
Object name is JCSM-13-55-g002.jpg

The aetiology of cancer cachexia. Chemotherapy (similarly to cancer) enhances the production of TNF‐α, and IL‐6, IL‐1; anorexia increases IL‐6 and TNF‐α; radiotherapy can enhance the circulating IL‐6 levels. The increase of these cytokines, along with decreased physical activity and deconditioning, results in muscle mass depletion.

Cachexia diagnosis

The cachexia syndrome is a continuum: It is preceded by a pre‐cachectic stage, in which metabolic changes anticipate weight loss, and evolves to refractory cachexia, which is an irreversible catabolic condition. 27 Cancer‐induced cachexia is diagnosed in patients who have experienced in the previous 6 months: an involuntary weight loss >5%, or a weight loss >2% if body mass index (BMI) is <20 kg/m2, or a weight loss >2% and signs of muscle depletion (sarcopenia). 27 Therefore, despite its complex aetiology and heterogeneous clinical features, the diagnosis of cachexia mainly relies on body weight. Weight loss is a well‐established negative prognostic factor for patients with cancer, but it cannot picture all the domains involved in the cachexia syndrome and it cannot distinguish between fat and muscle mass loss. Most importantly, weight loss cannot discriminate pre‐cachectic from non‐cachectic patients, not allowing an early identification of cachectic patients and prompt intervention. 28 Weight loss cut‐offs used by this definition have been validated in a cohort of patients with various cancer types. These have a prognostic value for cachectic vs. non‐cachectic patients (median OS 139 days vs. 269 days respectively; P < 0.001, n = 861), but not for pre‐cachectic vs. cachectic patients. Moreover, 56% of the patients enrolled were inpatients and the median Karnofsky Performance Status (KPS) was 70%, thus jeopardizing the utility of these cut‐offs for outpatients recently diagnosed with cancer or for those with a good KPS. 28

Sarcopenia—the low skeletal muscle mass and muscle strength—caused by the cachexia syndrome can anticipate the weight loss and can occur even in patients with overweight. 29 Therefore, diagnostic tools for sarcopenia should not rely on body weight. The gold standard diagnosis of sarcopenia is a two‐step approach 29 : first measuring handgrip strength and, if low, assessing the appendicular muscle mass (ASM), which constitutes the four limbs muscle mass to confirm the sarcopenia diagnosis. 30 In Table 1 , the cut‐offs for the diagnosis of sarcopenia are presented, along with alternative and easier to perform measurements.

Table 1

Most common diagnostic test for sarcopenia

MeasureTestSarcopenia diagnostic cut‐offs
Handgrip strengthHand‐held hydraulic dynamometer 31 <27 kg in men, <16 kg in women 32
ASMDEXA,BIA 29 <20 kg in men, <15 kg in women 30
ASM/height2 DEXA,BIA<7.0 kg/m2 in men, <5.5 kg/m2 in women 33
L3SMICT scan <52.4 cm 2 /m 2 in men, <38.5 cm 2 /m 2 in women 34
L3SMI CT scan <53 cm 2 /m 2 or <43 cm 2 /m 2 if BMI <25 kg/m 2 in men, <41 cm 2 /m 2 in women
L1SMI 35 CT scanTo be validated
Gait speed≤0.8 m/s 29
Timed up and go test≥20 s 29

Abbreviations: ASM, appendicular muscle mass; BIA, bioelectrical impedance analysis; DEXA, dual‐energy X‐ray absorptiometry; L1SMI, L1 skeletal muscle index, defined as the muscle cross‐sectional area (CSA) at L3 (cm2) divided by the height squared (m2); L3SMI, L3 skeletal muscle index, defined as the muscle cross‐sectional area (CSA) at L3 (cm2) divided by the height squared (m2).

Why is cachexia a problem in patients with lung cancer?

Cachexia occurs in about 40% of patients with metastatic NSCLC 6 and in approximately 20% of patients with early stage lung cancer. 14 In patients with advanced lung cancer, cachexia at diagnosis is associated with shorter survival (n = 226, median OS 11 vs. 6 months, P = 0.03). 14 Muscle depletion represents a negative prognostic factor for patients with unresectable NSCLC (n = 936) undergoing chemo‐radiotherapy with curative intent. 36 Cachexia develops and negatively affects the prognosis also in early‐stage cancer, even though the caloric intake is still preserved. This has been shown in non‐metastatic breast cancer [n = 3,241, hazard ratio (HR) for mortality 1.41; 95% confidence interval (CI): 1.18–1.69] 37 ; in Stages I–II colorectal cancer (n = 974, HR 2.22; 95% CI: 1.06–4.68) 38 and in early‐stage NSCLC (n = 90, 5 year‐survival: 72.8% vs. 85.8%, P = 0.028 39 ; n = 215 median OS 32 vs. 112 months). 40 The negative prognostic value of cachexia has also been shown in patients with NSCLC treated with immunotherapy (Table 2 ). Cachexia may undermine the immune system and increase the circulating levels of glucocorticoids, hindering the efficacy of immune checkpoint inhibitors (ICI), but this hypothesis has not yet been confirmed. 41

Table 2

The prognostic value of cachexia and sarcopenia during immune therapy

ICI (setting) N Study designCachexia/sarcopenia measureEndpointResults (non‐cachectic vs. cachectic pts)
Anti PD(L)‐1 (advanced NSCLC) 41 142Retrospective>5% weight loss in the previous 6mo.
  • DCR
  • PFS
  • OS
  • 59.9% vs. 41%, OR 2.60 (95% CI: 1–6.58)
  • Non statistically different
  • HR 6.26 (95% CI: 2.23–17.57)
Anti CTLA‐4 (advanced melanoma) 42 97RetrospectiveCT SMD < 42HU if BMI < 25 kg/m2 and <20 HU if BMI ≥ 25 kg/m2
  • PFS
  • OS
  • 2.7 vs. 2.4 months
  • 17.5 vs. 5.4 months

Anti PD(L)‐1

(various cancers) 43

100RetrospectiveL3SMI by CT (ROC cut‐offs)
  • PFS
  • OS
  • 7.5 months (95% CI: 2.9–10.9) vs. 3.3 months (95% CI: 2.8–5)
  • 15.6 months (95% CI: 12–21.9) vs. 4.7 months (95% CI: 4.1–6.6)

Atezolizumab

(Advanced NSCLC) 44

1,434Post‐hoc pooled analysis (4 phase III RCTs)BMI (18–24.9 vs. 25–29.9 vs. >30)
  • PFS
  • OS (BMI 25.0–29.9 vs. BMI < 25)
  • OS (BMI > 30 vs. BMI < 25)
  • NS
  • HR: 0.81 (95% CI: 0.68–0.95)
  • HR 0.64 (95% CI: 0.51–0.81)
Anti PD(L)‐1 (advanced NSCLC) 45 576Systematic meta‐analysis (9 RCTs)L3SMI, L3PMI.
  • PFS
  • % irAEs
  • OS
  • HR = 1.98 (95% CI: 1.32–2.97)
  • RR = 0.99 (95% CI: 0.21–4.67)
  • HR = 1.61 (95% CI: 1.24–2.10)

Abbreviations: BMI, body mass index; CTLA‐4, cytotoxic T‐lymphocyte antigen 4; DCR, disease control rate; HR, hazard ratio; HU, Hounsfield unit; irAEs, immune‐related adverse events; L3PMI, psoas muscle area at L3; L3SMI, skeletal muscle index at L3vertebra; NS, not significant; NSCLC, non‐small cell lung cancer; OS, overall survival; PD(L)‐1, programmed death ligand‐1; PFS, progression free survival; ROC, receiving operator curve; RR, risk ratio; SMD, skeletal muscle radiographic density.

The knowledge gap: what we do not know

Cachexia's negative prognostic impact is well established, but there are no reliable data about its predictive value for anticancer treatment, because all the published studies evaluating the association between cachexia and anticancer treatment were retrospective. Multiple factors, such as tumour histology and molecular profile, are predictive for anticancer treatment, and it is difficult to evaluate whether (pre)cachexia also has a negative predictive value, besides its known negative prognostic impact. The presence of cachexia (and sarcopenia) is not systematically addressed in clinical trials, and tumours that induce cachexia cannot be distinguished from a seemingly clinically similar one (e.g. stage, histology, and molecular analysis), that has no systemic impact. Consequently, the response to anti‐cancer treatments may be related to the tumour characteristics and not to the presence of cachexia. 46

Furthermore, the currently used definitions of cancer cachexia and sarcopenia have some limits. Rigid thresholds are used for a cachexia diagnosis, despite being described as a continuum. 27 Thresholds may differ according to the type of cancer and type of treatment and continuous variables instead of ordinal measures may help in identifying pre‐cachectic patients. Alternative scores include other domains associated with cachexia (immunosuppression, inflammation, QoL, loss of appetite, and daily activities). However, because of their complexity and variable relation with survival, they are not recommended in clinical practice. 47 Regarding sarcopenia, clinical trials assessed muscle performance through different functional tests (e.g. handgrip test, chair test, 6 min walking test), leading to heterogeneous results. Depending on the test used, patients may be diagnosed with or without cachexia or sarcopenia. 48 More importantly, we do not know whether an improvement in physical functioning would translate to better QoL and survival.

At present, there is no established treatment for cachexia in patients with cancer and the benefits of solely nutritional supplementation on cancer cachexia are evident only for patients with hypo‐nutrition as a consequence of mechanical obstacles, such as esophagitis‐induced dysphagia. 18 Furthermore, different combinations of muscle‐wasting triggers (anorexia, chemotherapy, deconditioning, and cancer itself) might imply different optimal cachexia treatments. 46 Physical exercise has a strong biological rationale for counteracting cancer‐cachexia and boosting the immune system (Figure 1 ), but the evidence is currently not sufficient to recommend it as cachexia treatment. 18 Furthermore, rationale and pre‐clinical evidences should be coupled with feasibility to translate the evidence into an appropriate intervention for patients with lung cancer.

Could physical exercise improve muscle mass and muscle performance in patients with cancer?

Physical exercise attenuates the age‐related physical decline in elderly and improves muscle mass, therefore it might counteract the muscle wasting induced by the cancer cachexia syndrome. 19

In a pre‐clinical study performed in colon carcinoma‐bearing mice, physical exercise attenuated body weight loss, muscle mass loss and increased appetite, compared to sedentary tumour bearing controls (P = 0.05). No differences were observed between sedentary and exercised healthy mice in terms of body weight. 49 Alves et al. showed that aerobic training improved running capacity and prolonged lifespan in Walker 256 rats bearing bone cancer. Aerobic interval activity also limited muscle mass loss and doubled the proteasome activity (responsible for protein quality control) within the plantaris muscle of exercised rats. 50 A 2020 systematic review of the literature (20 pre‐clinical trials) did not find any differences between exercised mice and controls regarding body mass nor muscle mass. Great heterogeneity among trials was reported (Q = 165.8, P < 0.01, τ2 = 6.1, I 2 = 95.5%). Training modalities (duration and intensity) and outcomes measures significantly differed among the trials. 51 In several studies involving tumour‐bearing mice, the training period started before the tumour inoculation; therefore, the fitness reached before the tumour inoculation, other than the exercise after the cancer‐inoculation, may have determined the results. 51

In patients with operable lung cancer (RCT, n = 151), pre‐operative supervised cycle‐ergometer training (2–3 times per week) improved the median peak oxygen consumption (+15%, IQR + 9–22%) and the median 6 min walking distance (6MWD) (+15%, IQR + 8–28%, P < 0.001). 52 A supervised exercise programme significantly mitigated the decline in VO2 peak, QoL and fatigue in patients with testicular‐, breast‐ and colon cancer during curative‐intent chemotherapy. 53 An RCT (n = 46) investigated the utility of endurance training (6 min walking in the hallway five times a week for 6 weeks plus strength exercise plus staircase exercises) in patients with advanced NSCLC during chemotherapy. Patients undergoing the programme showed an improvement in physical independence but general QoL did not differ. 54 Similarly, Cheville et al. showed that 8 weeks of home‐based endurance training improved mobility SF (P = 0.01) and FACT‐F (P = 0.02) QoL scores in patients with stage IV NSCLC or colorectal cancer. However, global QoL and survival did not differ. 55 A 2019 Cochrane review (6 RCTs, n = 221) highlighted the low quality of scientific evidence about exercise in patients with advanced lung cancer. The 6MWD (3 RCTs, n = 59) was significantly higher in the exercised group, but CIs were extremely wide (+63.33 m; 95% CI: 3.70–122.96), questioning the clinical meaning of this finding. No differences were found in muscle strength, dyspnoea, and fatigue. 56 In 41 patients with advanced cancer, a 12 weeks training programme (electrical muscle stimulation and strength exercise, performed twice a week for 20 min) significantly improved 6MWD (P = 0.006), skeletal muscle mass (P = 0.03) and body weight (P = 0.003). 57 131 patients—15.6% with NSCLC—with advanced cancer (non‐randomized CT) received nutritional support only or combined with a supervised exercise programme two times per week for 12 weeks. The experimental group had a higher mean skeletal muscle mass assessed with bioelectrical impedance analysis (+0.53 kg, 95% CI: 0.08–0.98) and mean body weight (+1.02 kg, 95% CI: 0.05–1.98). Fat mass did not differ between the two groups. The training improved 6MWD (P = 0.037) and KPS (P = 0.025). 58 Combined, these studies suggest that physical exercise in patients with (lung) cancer is feasible and play a role in promoting muscle anabolism and performance.

Does physical exercise improve quality of life and survival?

Physical exercise has been associated with health benefits in several conditions characterized by chronic inflammation. In patients with COPD, walking or cycling highly reduces symptoms and acute dyspnoea episodes. 59 In patients with human immunodeficiency virus (HIV), aerobic exercise (2–3 sessions per week, 30–90 min each) increases both cardiovascular fitness and QoL (systematic meta‐analysis, 7 RCTs). 60 Three training sessions/week of 30 min each (80–90% VO2max) in patients with rheumatoid arthritis led to a 38% decrease in disease activity according to the DAS28 score (n = 12). No changes in the circulating cytokines pre‐intervention and post‐intervention were reported. 61 In ankylosing spondylitis patients (n = 24), a reduction in disease activity was observed following 12 weeks of endurance training. 62

In a breast tumour‐bearing mouse model, 8 weeks of aerobic training increased the CD8+/FoxP3 + Treg circulating cells ratio, slowed tumour growth, and improved survival. Intriguingly, no differences in survival and tumour growth were seen in nude mice (exercised vs. sedentary) implying that the physical exercise effect on survival was mediated by the immune system. 63

Alves et al. showed that in tumour‐bearing mice, short‐term high‐intensity running could slow tumour progression and improve survival. 64 Contradictory results were also reported: Survival and tumour growth were not affected in mice with tumour‐induced cachexia practicing resistance training (climbing). 65

A higher weekly physical activity (90th percentile of physical activity compared with 10th percentile) reduced the incidence of various cancer, including lung cancer (HR = 0.74, 95% CI: 0.71–0.77), irrespective of BMI and smoking status (n = 1.44 mil.). 66 In the non‐metastatic setting, a post‐diagnosis higher level of physical exercise was associated with lower cancer‐specific mortality. However, causality between increased physical activity and reduced tumour growth has not been investigated. Moreover, the most active group was compared with the less active group, but minimal protective physical activity levels were not investigated. 67

In an RCT (exercise data already described above), including patients with lung cancer, the improvement in the peak oxygen consumption and 6MWD did not translate into less postoperative adverse events (primary endpoint) but the incidence of pulmonary complications (23% vs. 44%, P = 0.018) and atelectasis (12.2% vs. 36.4%, P < 0.001) decreased. 52

A 60 min supervised exercise programme (strength and balance exercises) twice a week for 3 months, combined with nutritional counselling session in patients with advanced cancer (RCT, n = 58, 30% NSCLC), did not show any improvement in global QoL (primary endpoint, assessed through EORT‐QLQ‐C30), and survival. 68 A Phase II RCT (n = 46) comparing multimodal intervention (nutritional supplements, FANS administration, and training programme) with standard care in patients with lung and pancreatic cancer did not find any differences in OS (secondary endpoint) (P = 0.57). 69 A single‐arm trial (n = 59) investigated the combination of nutrition counselling, fish oil supplementation, NSAID administrations, and physical exercise (9 weeks of twice per week lower body strength exercise and brisk‐pace walking) in patients with metastatic NSCLC. A gain in skeletal muscle was associated with treatment response, but causality was not investigated. 70 A supervised exercise programme, comprising cardiovascular and resistance training for a total of 9 h/week for 6 weeks during chemotherapy, both in the adjuvant and metastatic setting, led to a statistically significant improvement in fatigue (P = 0.02) and emotional well‐being. However, global QoL did not improve (RCT, n = 269). 71 Similarly, Oldervoll et al. (RCT, n = 231) found that a physical exercise intervention (60 min of circuit training twice a week for 8 weeks) improved handgrip strength but did not affect QoL. 72 Taken these studies together, there currently is a lack of high‐quality evidence that physical activity translates into a survival benefit. This is likely due to the small sample sizes and heterogeneity of patients included in the trials.

Do we know the most beneficial exercise intensity and duration?

The World Health Organization (WHO) recommends for all adults, also those affected by chronic conditions, 150–300 min of moderate‐intensity (Table 3)3) aerobic physical activity per week and resistance training of major muscle groups twice a week. 73 The principal investigated exercise schedules in patients with cancer comprise 2–3 sessions per week of 30–90 min‐aerobic or resistance training. A session >60 min was correlated with an improvement in lower and upper body muscle strength and supervised exercise appears to improve muscle strength and aerobic fitness (in terms of peak oxygen uptake). 74 The clinical meaning of these findings is not known. In patients receiving adjuvant therapy for breast cancer (RCT, n = 301) 60 min sessions (three times per week) of aerobic exercise are superior to 30 min sessions in reducing pain (P = 0.02), endocrine symptoms (P = 0.02), and in improving physical functionality, assessed trough Medical Outcomes Survey‐Short Form (SF)‐36 (P = 0.04). 75 A systematic meta‐analysis (n = 136 CTs) presents a non‐linear dose–response association between post‐diagnosis physical activity and all‐cause mortality in breast cancer survivors. Five, 10, 20, 30, and 65 MET‐hours per week were associated with an all‐cause mortality reduction of 22%, 43%, 59%, 69%, and 108%, respectively. The steep improvement in survival was lower for physical activity >15 MET‐hours per week. 76 However, this association does not imply causality. A single bout of 45 min of moderate‐intensity aerobic training increased muscle blood flow and amino acid delivery to the muscle in the elderly (70 ± 3 years, n = 6), regardless of the overall weekly physical activity. 77 Aerobic moderate‐intensity training (cycle‐ergometer) increased the volume of the quadriceps muscle in young (n = 7; mean age 20 ± 1 years) and older adults (n = 6; mean age 74 ± 3 years) by a similar amount, despite the elderly group completing half of the mechanical work. 78 Young men (median age 24.8 years) and older men (median age 70 years) received the same volume of resistance training for 12 weeks (n = 38). Older men experienced a significantly greater increase in triceps muscle volume, assessed through MRI (P < 0.05). 79 These findings suggest that the basal muscle status determines the volume of exercise needed to improve muscle mass. 80 In summary, the optimal exercise volume and type needed for patients with cancer is currently unknown. Moreover, the most crucial parameter for a training programme effective in counteracting cachexia might not be the total amount of physical activity but its increase compared with baseline. Tailoring the physical activity on the patients' attitude and basal fitness would also improve compliance, considering that patients with cancer cannot be expected to train at the same intensity levels of a healthy individual.

Table 3

Exercise intensity definitions 72

Metabolic equivalent of task (MET)The ratio of the energy used up during an activity divided by the energy expended at rest (e.g. a 4 MET activity requires four times more energy than at rest).
Aerobic exercise (also named endurance activity)Activity in which large muscles are used in a rhythmic manner. During aerobic activity, the body produce energy using oxygen (e.g. bicycling, walking, and running).
Resistance exerciseMuscles contracts and work against a force or a weight (e.g. heavy lifting, push‐ups).
Light intensity physical activityActivity performed between 1.5 and 3 METs, without increasing heart rate (e.g. walking at a slow pace).
Moderate intensity activityActivity performed between 3 and 6 METs.
Vigorous physical activityActivity performed at >6.0 METs.

Can physical exercise counteract chronic inflammation?

In healthy humans (CT, n = 16) injected with Escherichia coli endotoxin, mimicking chronic inflammation, TNF‐α was increased by two‐fold to three‐fold in the resting cohort. In contrast, TNF‐α did not increase in subjects performing 3 h of cycling at 75% VO2 peak after the injection. 81 Physical exercise protects from chronic inflammation avoiding an increase in visceral fat. A reduction from 10 000 steps per day to 1500 steps per day for 14 days in healthy individuals increased abdominal visceral fat while reducing BMI and lean mass. 82 A 3 months exercise programme in obese individuals led to weight and visceral fat reduction, which lasted even 12 months after the intervention. Subjects achieving the same weight loss through dietary restriction alone re‐gained the visceral fat at 12 months. 83 A meta‐analysis (RCT and CT = 83, n = 3769) showed that physical exercise reduced C‐reactive protein (used as a proxy of systemic inflammation). The decrease in CRP was associated with decreased BMI and percentage of visceral fat. 84 An RCT performed in patients with breast cancer receiving adjuvant radiotherapy (n = 103) showed that IL‐6 was increased in the sedentary group during treatment, while no change was noted in the patients undergoing physical exercise. Fatigue and pain, measured through EORTC‐ QLQ‐C30, were also significantly reduced in the exercised group. 85 IFN‐γ decreased significantly in patients with non‐metastatic cancer (n = 292) who received chemotherapy and a 6 week training programme (prescribed number of daily steps, which increases 5–20% per week, and completing a prescribed number of sets and repetitions of resistance exercises), compared to chemotherapy alone. However, IL‐6 and IL‐10 levels did not differ. 86

Physical exercise might also lower systemic inflammation by enhancing the metabolism of mitochondria, consequently decreasing mitochondrial reactive oxygen species (ROS) production. In age‐associated sarcopenia, mitochondria appear to be reduced and dysfunctional, thus leading to an increased accumulation of ROS within the muscles. The ROS increase ultimately promotes local and systemic inflammation, as observed in obese individuals. 87 In mice, treadmill running has shown to decrease muscle ROS accumulation. 88 Twelve weeks of resistance training has shown to lower oxidative stress also within the muscles of elderly subjects. 89 The implications of mitochondria metabolism for immune activity against cancer cells need to be further investigated.

The reduction of systemic chronic inflammation mediated by physical exercise may also translate into a more effective host immune system.

Can physical exercise boost the immune system?

Muscle cells can secrete over 600 myokines, exerting paracrine and endocrine effects. Muscle cells also release IL‐6, IL‐7, and IL‐15 influencing the immune environment, both locally and at a systemic level. 9 IL‐15 promotes muscle cell regeneration, reduces adiposity and expands and activates natural killer (NK) and CD8 + T cells. 90 In rodents, 45 min exercise bouts promote leukocytes' recirculation between peripheral tissues (lung, spleen, and muscle) and the bloodstream. 91 Batista et al. investigated exercise training in rats with heart failure (which is associated with a change in skeletal muscle, possibly due to chronic inflammation after heart failure). After an 8 week programme, the trained rats had lower serum levels of TNF‐α protein (−26%, P < 0.05) and lower levels of TNF‐α mRNA within the soleus muscle, while IL‐10 was 2.6‐fold higher (P < 0.001). 92 In breast cancer‐bearing mice, physical exercise increased the NK lymphocytes infiltrating tumour tissue 93 and lowered intra‐tumour myeloid‐derived suppressor cells. 94 IL‐15 serum levels increase in humans in response to physical exercise. 95 In a Phase‐I clinical trial performed in patients with melanoma (n = 5), recombinant IL‐15 injection promoted NK lymphocytes and CD8 + T cells mobilization from the blood into peripheral tissue. 96 Low IL‐7 levels are associated with a senescent immune system 97 and physical exercise in the elderly (n = 255) maintains adequate IL‐7 levels. 98 During training, IL‐6 is secreted by muscle pulsatile in the absence of TNF‐α and promotes muscle anabolism. 11 Physically active subjects present higher levels of circulating CD8 + lymphocytes and lower levels of IL‐6 compared with sedentary subjects. 98 In healthy individuals, IL‐6 levels are increased in response to exercise. The most critical factor for IL‐6 increase is exercise duration. 99 After 30 min of running at 75% VO2 peak, IL‐6 increased five‐fold 100 while it increased to 8000‐fold, after an extreme effort, such as running for 246 km. 101 IL‐6 increases exponentially during exercise, reaching the peak at the end of the training and rapidly decreasing afterward. 100 The intensity of the exercise also influences IL‐6: in marathon runners (n = 53), IL‐6 increase was directly correlated with running intensity (calculated as running speed divided by VO2 max). 102 IL‐6 mRNA in the human vastus lateralis muscle, assessed through biopsies, increased up to 100‐fold at the end of the training (n = 6). 103 The same study showed how TNF‐a mRNA did not increase during exercise. 103 Injection of recombinant human IL‐6, mimicking exercise‐secreted IL‐6, increased IL‐10 and IL‐1 receptor agonist (IL‐1ra) in healthy individuals (placebo‐controlled trial, n = 12). 104 In patients with HIV (n = 30), physical exercise reduced the levels of circulating IL‐6, TNF‐a and IL‐1, while increasing CD8+ and IL‐10 levels. 105 Physical activity prevents immunosenescence during aging and improves immune response to vaccinations. 106 Importantly, the majority of the findings on how the exercise influences the immune system are derived from healthy individuals and are valid for certain physical‐activity thresholds: We do not know whether the same mechanisms would still work in patients with cancer and whether they would improve the patients' outcomes.

Intestinal microbiota: the missing piece of the puzzle?

The role of the intestinal bacteria—the so called gut microbiota—in influencing the interplay between the immune system and cancer is an emerging topic, and it might also influence the cancer‐cachexia development. The mechanisms by which the intestinal microbiome can influence immune system and muscle metabolism are still under investigation. Lipopolysaccharide (LPS) is a major component of gram‐negative bacteria and can elicit inflammation by binding to extracellular toll‐like receptor‐4 (TLR‐4). Continuous translocations of LPS from intestinal wall into bloodstream can induce chronic inflammation. A higher proportion of gram‐positive microorganisms, such as Lactobacillus, within the microbiome might lower the proliferation rate of gram‐negative microorganisms and consequently, lower LPS production. Exercise training has also shown to lower LPS blood levels in healthy individuals. 107 Consistently in mice, a lower proportion of bacterioides (gram negative) in the intestinal microbiome has been correlated with lower systemic inflammation. 108 The microbiome diversity and the Lactobacillus spp. were reduced in mice models of cancer‐induced cachexia compared to healthy controls. Moreover, restoring the Lactobacillus spp. by oral supplementation led to a reduction of atrophy markers (MuRF1, Atrogin‐1, LC3, and Cathepsin L) in the gastrocnemius muscle, along with a reduction in circulating IL‐6. 109 A subsequent study, performed in leukaemic mice, confirmed that Lactobacillus population was restored by administering live Lactobacillus reuteri , and this translated into a diminished cancer cell proliferation, an attenuated muscle wasting and an increased survival. 110 Butyrate is a short‐chain fatty acid, which can suppress the activation of NF‐κB and inhibit IFN‐γ signalling, thus reducing chronic inflammation and is mainly produced the Ruminococcaceae within the intestinal microbiome. 111 The diversity of the gut microbiome along with high levels of Ruminococcaceae and Agathobacter have been associated with better survival in patients with cancer. 112 Pötgens et al. showed that in cachectic colon cancer bearing mice the levels of Ruminococcaceae were reduced compared to healthy controls. 113 Moreover, preliminary studies suggest that microbiome diversity might even enhance immunotherapy response in patients with cancer. 114 In mice with depleted intestinal microbiota after broad‐spectrum antibiotics treatment, the running capacity and the muscle contractile capacity were decreased. After the natural restoration of microbiota, the physical performance was also normalized. The antibiotics exposure altered the gene expression of glucose transporters in the ileum and the glycogen levels in the muscle, suggesting that the gut microbiome might influence the muscle performance also through the glucose/glycogen metabolism. 115 The relation between intestinal microbiome and physical activity seems to work also the other way around: in healthy individuals (n = 41) the cardiorespiratory fitness—estimated through VO2 peak—predicted microbiome diversity. 116 Moreover, intestinal microbiome diversity has been found to be increased in professional rugby players compared with healthy and BMI‐matched controls (n = 86). 117 Physical activity in healthy individuals has not only shown to increase gut microbiota diversity but also to enhance benign micro‐organisms levels, such as Ruminococcaceae. 117 Investigating the link between nutrition, physical exercise, the microbiome and the immune system would be of the utmost importance in the next years in order to reach an actual personalized immunotherapy treatment for patients with cancer.

Future perspectives

We need a deeper understanding of cachexia development and its relation with the immune system to provide adequate interventions to patients with cancer, especially as ICI or other immunotherapeutic agents are, or will become, standard of care in multiple histology. Future trials should dynamically follow the evolution of cancer cachexia and immune system changes during cancer treatment (e.g. with circulating biomarkers or muscle mass CSA measurements). Patients with pre‐cachexia, cachexia, and refractory cachexia, should not be mixed in clinical trials, in order to properly evaluate the efficacy of novel interventions and to achieve a better mechanistic understanding of the cachexia process. Nutrition and physical exercise should be investigated agnostically, considering that they might also be detrimental. We do not know if boosting the immune system through exercise could result in hyper‐progression, as has been described for ICI treatment. Trials should stratify patients according to different cachexia mechanisms and tumour characteristics, aiming to achieve a personalized cachexia treatment, similar to for example oncogene‐addicted NSCLC.

Methods

In the present narrative review, we included animal models of cancer cachexia that investigated whether exercise or nutrition would improve muscle mass or muscle performance; whether exercise would influence the immune system; whether exercise would provide a survival benefit; whether exercise would influence tumour growth. Animal models were included if their findings were mechanistically relevant to explain the effect of physical exercise on human muscle and immune system. We did not include clinical trials in humans investigating solely the feasibility of an exercise programme. Furthermore, trials were not included if only the abstract was available or the manuscript's full text was not available in English.

Conflict of interest

This research did not receive any specific grant from funding agencies in the public, commercial, or not‐for‐profit sectors. Dirk De Ruysscher has received a grant paid to the institution from AstraZeneca, no personal fees. Lizza Hendriks has received grants paid to the institution from Roche Genentech, Boehringer Ingelheim, Takeda, and AstraZeneca; honoraria paid to the institutions from MSD; support for attending meetings and/or travel from Roche Genentech; advisory board honoraria from BMS, Eli Lilly, Roche Genentech, Pfizer, Takeda, MSD, Boehringer Ingelheim, Amgen, and Janssen (all paid to the Institution except from Roche Genentech); mentorship programme and educational webinars fees from Medtalks, Benecke (self), Roche Genentech, and Bayer (paid to the Institution). Francesco Cortiula, Wouter van de Worp, Annemie Schols, Rianne Vaes, and Ramon Langen have no conflicts of interest to disclose.

Ethics statement

All authors comply with the Ethical guidelines for authorship and publishing in the Journal of Cachexia, Sarcopenia and Muscle. 118 The manuscript does not contain patient data.

Artwork

The artwork present in the manuscript has been prepared with Adobe Illustrator 2020®.

Acknowledgements

This Research Project was supported by the European Society for Medical Oncology (ESMO). Any views, opinions, findings, conclusions, or recommendations expressed in this material are those solely of the author(s) and do not necessarily reflect those of ESMO.

Notes

Cortiula F., Hendriks L. E. L., van de Worp W. R. P. H., Schols A. M. W. J., Vaes R. D. W., Langen R. C. J., and De Ruysscher D. (2022) Physical exercise at the crossroad between muscle wasting and the immune system: implications for lung cancer cachexia, Journal of Cachexia, Sarcopenia and Muscle, 13, 55–67, 10.1002/jcsm.12900 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

References

1. Evans WJ, Morley JE, Argilés J, Bales C, Baracos V, Guttridge D, et al. Cachexia: a new definition. Clin Nutr 2008;27:793–799. [PubMed] [Google Scholar]
2. Nipp R, Fuchs G, El‐Jawahri A, Mario J, Troschel F, Greer J, et al. Sarcopenia is associated with quality of life and depression in patients with advanced cancer. Oncologist 2018;23:97–104. [PMC free article] [PubMed] [Google Scholar]
3. Kwan HY, Maddocks M, Nolan CM, Jones SE, Patel S, Barker RE, et al. The prognostic significance of weight loss in chronic obstructive pulmonary disease‐related cachexia: a prospective cohort study. J Cachexia Sarcopenia Muscle 2019;10:1330–1338. [PMC free article] [PubMed] [Google Scholar]
4. Santo RCE, Fernandes KZ, Lora PS, Filippin LI, Xavier RM. Prevalence of rheumatoid cachexia in rheumatoid arthritis: a systematic review and meta‐analysis. J Cachexia Sarcopenia Muscle 2018;9:816–825. [PMC free article] [PubMed] [Google Scholar]
5. Mak RH, Cheung W. Energy homeostasis and cachexia in chronic kidney disease. Pediatr Nephrol 2006;21:1807–1814. [PubMed] [Google Scholar]
6. Anker MS, Holcomb R, Muscaritoli M, von Haehling S, Haverkamp W, Jatoi A, et al. Orphan disease status of cancer cachexia in the USA and in the European Union: a systematic review. J Cachexia Sarcopenia Muscle 2019;10:22–34. [PMC free article] [PubMed] [Google Scholar]
7. Langen RCJ, Haegens A, Vernooy JHJ, Wouters EFM, de Winther MPJ, Carlsen H, et al. NF‐κB activation is required for the transition of pulmonary inflammation to muscle atrophy. Am J Respir Cell Mol Biol 2012;47:288–297. [PMC free article] [PubMed] [Google Scholar]
8. Jones SA, Jenkins BJ. Recent insights into targeting the IL‐6 cytokine family in inflammatory diseases and cancer. Nat Rev Immunol 2018;18:773–789. [PubMed] [Google Scholar]
9. Nelke C, Dziewas R, Minnerup J, Meuth SG, Ruck T. Skeletal muscle as potential central link between sarcopenia and immune senescence. EBioMedicine 2019;49:381–388. [PMC free article] [PubMed] [Google Scholar]
10. Webster JM, Kempen LJAP, Hardy RS, Langen RCJ. Inflammation and skeletal muscle wasting during cachexia. Front Physiol 2020;11. [PMC free article] [PubMed] [Google Scholar]
11. Hartwig S, Raschke S, Knebel B, Scheler M, Irmler M, Passlack W, et al. Secretome profiling of primary human skeletal muscle cells. Biochim Biophys Acta 2014;1844:1011–1017. [PubMed] [Google Scholar]
12. Kaneki M. Metabolic inflammatory complex in sepsis: septic cachexia as a novel potential therapeutic target. Shock 2017;48:600–609. [PMC free article] [PubMed] [Google Scholar]
13. Op den Kamp CM, Langen RC, Minnaard R, Kelders MC, Snepvangers FJ, Hesselink MK, et al. Pre‐cachexia in patients with stages I–III non‐small cell lung cancer: systemic inflammation and functional impairment without activation of skeletal muscle ubiquitin proteasome system. Lung Cancer 2012;76:112–117. [PubMed] [Google Scholar]
14. Lau S, Giap F, Gannavarapu BS, Iyengar P. Impact of cachexia at diagnosis on radiotherapy utilization and survival in non‐small cell lung cancer. JCO 2016;34:133–133. [Google Scholar]
15. Op den Kamp CMH, De Ruysscher DKM, van den Heuvel M, Elferink M, Houben RMA, Oberije CJG, et al. Early body weight loss during concurrent chemo‐radiotherapy for non‐small cell lung cancer. J Cachexia Sarcopenia Muscle 2014;5:127–137. [PMC free article] [PubMed] [Google Scholar]
16. Sadeghi M, Keshavarz‐Fathi M, Baracos V, Arends J, Mahmoudi M, Rezaei N. Cancer cachexia: diagnosis, assessment, and treatment. Crit Rev Oncol Hematol 2018;127:91–104. [PubMed] [Google Scholar]
17. Lowell BB, Goodman MN. Protein sparing in skeletal muscle during prolonged starvation: dependence on lipid fuel availability. Diabetes 1987;36:14–19. [PubMed] [Google Scholar]
18. Roeland EJ, Bohlke K, Baracos VE, Bruera E, Del Fabbro E, Dixon S, et al. Management of cancer cachexia: ASCO guideline. J Clin Oncol 2020;38:2438–2453. [PubMed] [Google Scholar]
19. Mendonca GV, Pezarat‐Correia P, Vaz JR, Silva L, Almeida ID, Heffernan KS. Impact of exercise training on physiological measures of physical fitness in the elderly. Curr Aging Sci 2016;9:240–259. [PubMed] [Google Scholar]
20. Yang M, Shen Y, Tan L, Li W. Prognostic value of sarcopenia in lung cancer: a systematic review and meta‐analysis. Chest 2019;156:101–111. [PubMed] [Google Scholar]
21. Gadgeel S, Rodríguez‐Abreu D, Speranza G, Esteban E, Felip E, Dómine M, et al. Updated analysis from KEYNOTE‐189: pembrolizumab or placebo plus pemetrexed and platinum for previously untreated metastatic nonsquamous non‐small‐cell lung cancer. J Clin Oncol 2020;38:1505–1517. [PubMed] [Google Scholar]
22. Gibson D, Mehler PS. Anorexia nervosa and the immune system—a narrative review. J Clin Med 2019;8. [PMC free article] [PubMed] [Google Scholar]
23. Damrauer JS, Stadler ME, Acharyya S, Baldwin AS, Couch ME, Guttridge DC. Chemotherapy‐induced muscle wasting: association with NF‐κB and cancer cachexia. Eur J Transl Myol 2018;28:7590. [PMC free article] [PubMed] [Google Scholar]
24. Stubblefield MD. Neuromuscular complications of radiation therapy. Muscle Nerve 2017;56:1031–1040. [PubMed] [Google Scholar]
25. D'Souza D, Roubos S, Larkin J, Lloyd J, Emmons R, Chen H, et al. The late effects of radiation therapy on skeletal muscle morphology and progenitor cell content are influenced by diet‐induced obesity and exercise training in male mice. Sci Rep 2019;9:6691. [PMC free article] [PubMed] [Google Scholar]
26. Convertino VA, Bloomfield SA, Greenleaf JE. An overview of the issues: physiological effects of bed rest and restricted physical activity. Med Sci Sports Exerc 1997;29:187–190. [PubMed] [Google Scholar]
27. Fearon K, Strasser F, Anker SD, Bosaeus I, Bruera E, Fainsinger RL, et al. Definition and classification of cancer cachexia: an international consensus. Lancet Oncol 2011;12(5):489–495. 10.1016/s1470-2045(10)70218-7 [PubMed] [CrossRef] [Google Scholar]
28. Blum D, Ts S, Solheim T, Hjermstad M, Kaasa S. Validation of the consensus‐definition for cancer cachexia and evaluation of a classification model—a study based on data from an international multicentre project (EPCRC‐CSA). Ann Oncol 2014;25:1635–1642. [PubMed] [Google Scholar]
29. Cruz‐Jentoft A, Bahat G, Bauer J, Boirie Y, Bruyère O, Cederholm T, et al. Sarcopenia: revised European consensus on definition and diagnosis. Age Ageing 2019;48:601. [PMC free article] [PubMed] [Google Scholar]
30. Studenski SA, Peters KW, Alley DE, Cawthon PM, McLean RR, Harris TB, et al. The FNIH sarcopenia project: rationale, study description, conference recommendations, and final estimates. J Gerontol A Biol Sci Med Sci 2014;69:547–558. [PMC free article] [PubMed] [Google Scholar]
31. Reijnierse EM, de Jong N, Trappenburg MC, Blauw GJ, Butler‐Browne G, Gapeyeva H, et al. Assessment of maximal handgrip strength: how many attempts are needed? J Cachexia Sarcopenia Muscle 2017;8:466–474. [PMC free article] [PubMed] [Google Scholar]
32. Dodds RM, Syddall HE, Cooper R, Benzeval M, Deary IJ, Dennison EM, et al. Grip strength across the life course: normative data from twelve British studies. PLoS ONE 2014;9:e113637. [PMC free article] [PubMed] [Google Scholar]
33. Gould H, Brennan SL, Kotowicz MA, Nicholson GC, Pasco JA. Total and appendicular lean mass reference ranges for Australian men and women: the Geelong osteoporosis study. Calcif Tissue Int 2014;94:363–372. [PubMed] [Google Scholar]
34. Prado CMM, Lieffers JR, McCargar LJ, Reiman T, Sawyer MB, Martin L, et al. Prevalence and clinical implications of sarcopenic obesity in patients with solid tumours of the respiratory and gastrointestinal tracts: a population‐based study. Lancet Oncol 2008;9:629–635. [PubMed] [Google Scholar]
35. Martin L, Birdsell L, MacDonald N, Reiman T, Clandinin MT, McCargar LJ, et al. Cancer cachexia in the age of obesity: skeletal muscle depletion is a powerful prognostic factor, independent of body mass indexCachexia in the Age of Obesity: Skeletal Muscle Depletion Is a Powerful Prognostic Factor, Independent of Body Mass Index. JCO 2013;31:1539–1547. [PubMed] [Google Scholar]
36. Burtin C, Bezuidenhout J, Sanders KJC, Dingemans A‐MC, Schols AMWJ, Peeters STH, et al. Handgrip weakness, low fat‐free mass, and overall survival in non‐small cell lung cancer treated with curative‐intent radiotherapy. J Cachexia Sarcopenia Muscle 2020;11:424–431. [PMC free article] [PubMed] [Google Scholar]
37. Caan BJ, Cespedes Feliciano EM, Prado CM, Alexeeff S, Kroenke CH, Bradshaw P, et al. Association of muscle and adiposity measured by computed tomography with survival in patients with nonmetastatic breast cancer. JAMA Oncol 2018;4:798–804. [PMC free article] [PubMed] [Google Scholar]
38. Shirdel M, Andersson F, Myte R, Axelsson J, Rutegård M, Blomqvist L, et al. Body composition measured by computed tomography is associated with colorectal cancer survival, also in early‐stage disease. Acta Oncol 2020;59:799–808. [PubMed] [Google Scholar]
39. Suzuki Y, Okamoto T, Fujishita T, Katsura M, Akamine T, Takamori S, et al. Clinical implications of sarcopenia in patients undergoing complete resection for early non‐small cell lung cancer. Lung Cancer 2016;101:92–97. [PubMed] [Google Scholar]
40. Tsukioka T, Nishiyama N, Izumi N, Mizuguchi S, Komatsu H, Okada S, et al. Sarcopenia is a novel poor prognostic factor in male patients with pathological Stage I non‐small cell lung cancer. Jpn J Clin Oncol 2017;47:363–368. [PubMed] [Google Scholar]
41. Flint TR, Janowitz T, Connell CM, Roberts EW, Denton AE, Coll AP, et al. Tumor‐induced IL‐6 reprograms host metabolism to suppress anti‐tumor immunity. Cell Metab 2016;24:672–684. [PMC free article] [PubMed] [Google Scholar]
42. Roch B, Coffy A, Jean‐Baptiste S, Palaysi E, Daures J‐P, Pujol J‐L, et al. Cachexia—sarcopenia as a determinant of disease control rate and survival in non‐small lung cancer patients receiving immune‐checkpoint inhibitors. Lung Cancer 2020;143:19–26. [PubMed] [Google Scholar]
43. Chu MP, Li Y, Ghosh S, Sass S, Smylie M, Walker J, et al. Body composition is prognostic and predictive of ipilimumab activity in metastatic melanoma. J Cachexia Sarcopenia Muscle 2020;11:748–755. [PMC free article] [PubMed] [Google Scholar]
44. Cortellini A, Bozzetti F, Palumbo P, Brocco D, Di Marino P, Tinari N, et al. Weighing the role of skeletal muscle mass and muscle density in cancer patients receiving PD‐1/PD‐L1 checkpoint inhibitors: a multicenter real‐life study. Sci Rep 2020;10:1456. [PMC free article] [PubMed] [Google Scholar]
45. Kichenadasse G, Miners JO, Mangoni AA, Rowland A, Hopkins AM, Sorich MJ. Association between body mass index and overall survival with immune checkpoint inhibitor therapy for advanced non‐small cell lung cancer. JAMA Oncol 2020;6:512–518. [PMC free article] [PubMed] [Google Scholar]
46. Willemsen ACH, Degens JHRJ, Baijens LWJ, Dingemans A‐MC, Hoeben A, Hoebers FJP, et al. Early loss of fat mass during chemoradiotherapy predicts overall survival in locally advanced squamous cell carcinoma of the lung, but not in locally advanced squamous cell carcinoma of the head and neck. Front Nutr 2020;7:600612. [PMC free article] [PubMed] [Google Scholar]
47. Argilés JM, Betancourt A, Guàrdia‐Olmos J, Peró‐Cebollero M, López‐Soriano FJ, Madeddu C, et al. Validation of the CAchexia SCOre (CASCO). Staging cancer patients: the use of miniCASCO as a simplified tool. Front Physiol 2017;8:8. [PMC free article] [PubMed] [Google Scholar]
48. Yee XS, Ng YS, Allen JC, Latib A, Tay EL, Abu Bakar HM, et al. Performance on sit‐to‐stand tests in relation to measures of functional fitness and sarcopenia diagnosis in community‐dwelling older adults. Eur Rev Aging Phys Act 2021;18:1. [PMC free article] [PubMed] [Google Scholar]
49. Ballarò R, Penna F, Pin F, Gómez‐Cabrera MC, Viña J, Costelli P. Moderate exercise improves experimental cancer cachexia by modulating the redox homeostasis. Cancers (Basel) 2019;11. [PMC free article] [PubMed] [Google Scholar]
50. Alves CRR, Das NW, de Almeida NR, Eichelberger EJ, Jannig PR, Voltarelli VA, et al. Exercise training reverses cancer‐induced oxidative stress and decrease in muscle COPS2/TRIP15/ALIEN. Mol Metab 2020;39:101012. 10.1016/j.molmet.2020.101012 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
51. Niels T, Tomanek A, Freitag N, Schumann M. Can exercise counteract cancer cachexia? A systematic literature review and meta‐analysis. Integr Cancer Ther 2020;19:153473542094041. [PMC free article] [PubMed] [Google Scholar]
52. Licker M, Karenovics W, Diaper J, Frésard I, Triponez F, Ellenberger C, et al. Short‐term preoperative high‐intensity interval training in patients awaiting lung cancer surgery: a randomized controlled trial. J Thorac Oncol 2017;12:323–333. [PubMed] [Google Scholar]
53. Meeting Library Effect of a tailored exercise intervention during or after chemotherapy on cardiovascular morbidity in cancer patients. n.d. https://meetinglibrary.asco.org/record/187836/abstract (accessed January 15, 2021).
54. Henke CC, Cabri J, Fricke L, Pankow W, Kandilakis G, Feyer PC, et al. Strength and endurance training in the treatment of lung cancer patients in stages IIIA/IIIB/IV. Support Care Cancer 2014;22:95–101. [PubMed] [Google Scholar]
55. Cheville AL, Kollasch J, Vandenberg J, Shen T, Grothey A, Gamble G, et al. A home‐based exercise program to improve function, fatigue, and sleep quality in patients with stage IV lung and colorectal cancer: a randomized controlled trial. J Pain Symptom Manag 2013;45:811–821. [PMC free article] [PubMed] [Google Scholar]
56. Peddle‐McIntyre CJ, Singh F, Thomas R, Newton RU, Galvão DA, Cavalheri V. Exercise training for advanced lung cancer. Cochrane Database Syst Rev 2019;2:CD012685, 10.1002/14651858.CD012685.pub2 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
57. Schink K, Gaßner H, Reljic D, Herrmann HJ, Kemmler W, Schwappacher R, et al. Assessment of gait parameters and physical function in patients with advanced cancer participating in a 12‐week exercise and nutrition programme: a controlled clinical trial. Eur J Cancer Care 2020;29:e13199. 10.1111/ecc.13199 [PubMed] [CrossRef] [Google Scholar]
58. Schink K, Herrmann HJ, Schwappacher R, Meyer J, Orlemann T, Waldmann E, et al. Effects of whole‐body electromyostimulation combined with individualized nutritional support on body composition in patients with advanced cancer: a controlled pilot trial. BMC Cancer 2018;18:886. [PMC free article] [PubMed] [Google Scholar]
59. He GX, Li N, Ren L, Shen HH, Liao N, Wen JJ, et al. Benefits of different intensities of pulmonary rehabilitation for patients with moderate‐to‐severe COPD according to the GOLD stage: a prospective, multicenter, single‐blinded, randomized, controlled trial. Int J Chron Obstruct Pulmon Dis 2019;14:2291–2304. [PMC free article] [PubMed] [Google Scholar]
60. Gomes Neto M, Conceição CS, Carvalho VO, Brites C. Effects of combined aerobic and resistance exercise on exercise capacity, muscle strength and quality of life in HIV‐infected patients: a systematic review and meta‐analysis. PLoS ONE 2015;10:e0138066. [PMC free article] [PubMed] [Google Scholar]
61. Bartlett DB, Willis LH, Slentz CA, Hoselton A, Kelly L, Huebner JL, et al. Ten weeks of high‐intensity interval walk training is associated with reduced disease activity and improved innate immune function in older adults with rheumatoid arthritis: a pilot study. Arthritis Res Ther 2018;20:127. [PMC free article] [PubMed] [Google Scholar]
62. Sveaas SH, Berg IJ, Provan SA, Semb AG, Hagen KB, Vøllestad N, et al. Efficacy of high intensity exercise on disease activity and cardiovascular risk in active axial spondyloarthritis: a randomized controlled pilot study. PLoS ONE 2014;9:e108688. [PMC free article] [PubMed] [Google Scholar]
63. Hagar A, Wang Z, Koyama S, Serrano JA, Melo L, Vargas S, et al. Endurance training slows breast tumor growth in mice by suppressing Treg cells recruitment to tumors. BMC Cancer 2019;19:536. [PMC free article] [PubMed] [Google Scholar]
64. Alves CRR, Das NW, Tobias GC, de Almeida NR, Barreto RF, Melo CM, et al. High‐intensity interval training slows down tumor progression in mice bearing Lewis lung carcinoma. JCSM Rapid Communications 2018;1:1–10. [Google Scholar]
65. das Neves W, Alves CRR, de Almeida NR, Guimarães FLR, Ramires PR, Brum PC, et al. Loss of strength capacity is associated with mortality, but resistance exercise training promotes only modest effects during cachexia progression. Life Sci 2016;163:11–22. [PubMed] [Google Scholar]
66. Moore SC, Lee I‐M, Weiderpass E, Campbell PT, Sampson JN, Kitahara CM, et al. Leisure‐time physical activity and risk of 26 types of cancer in 1.44 million adults. JAMA Intern Med 2016;176:816–825. [PMC free article] [PubMed] [Google Scholar]
67. Friedenreich CM, Neilson HK, Farris MS, Courneya KS. Physical activity and cancer outcomes: a precision medicine approach. Clin Cancer Res 2016;22:4766–4775. [PubMed] [Google Scholar]
68. Uster A, Ruehlin M, Mey S, Gisi D, Knols R, Imoberdorf R, et al. Effects of nutrition and physical exercise intervention in palliative cancer patients: a randomized controlled trial. Clin Nutr 2018;37:1202–1209. [PubMed] [Google Scholar]
69. Solheim TS, Laird BJA, Balstad TR, Stene GB, Bye A, Johns N, et al. A randomized phase II feasibility trial of a multimodal intervention for the management of cachexia in lung and pancreatic cancer. J Cachexia Sarcopenia Muscle 2017;8:778–788. [PMC free article] [PubMed] [Google Scholar]
70. Tobberup R, Carus A, Rasmussen HH, Falkmer UG, Jorgensen MG, Schmidt EB, et al. Feasibility of a multimodal intervention on malnutrition in patients with lung cancer during primary anti‐neoplastic treatment. Clin Nutr 2020;40:525–533. [PubMed] [Google Scholar]
71. Adamsen L, Quist M, Andersen C, Møller T, Herrstedt J, Kronborg D, et al. Effect of a multimodal high intensity exercise intervention in cancer patients undergoing chemotherapy: randomised controlled trial. BMJ 2009;339:b3410. [PMC free article] [PubMed] [Google Scholar]
72. Oldervoll LM, Loge JH, Lydersen S, Paltiel H, Asp MB, Nygaard UV, et al. Physical exercise for cancer patients with advanced disease: a randomized controlled trial. Oncologist 2011;16:1649–1657. [PMC free article] [PubMed] [Google Scholar]
73. Bull FC, Al‐Ansari SS, Biddle S, Borodulin K, Buman MP, Cardon G, et al. World Health Organization 2020 guidelines on physical activity and sedentary behaviour. Br J Sports Med 2020;54:1451–1462. [PMC free article] [PubMed] [Google Scholar]
74. Sweegers MG, Altenburg TM, Brug J, May AM, van Vulpen JK, Aaronson NK, et al. Effects and moderators of exercise on muscle strength, muscle function and aerobic fitness in patients with cancer: a meta‐analysis of individual patient data. Br J Sports Med 2019;53:812–812. [PubMed] [Google Scholar]
75. Courneya KS, McKenzie DC, Mackey JR, Gelmon K, Friedenreich CM, Yasui Y, et al. Effects of exercise dose and type during breast cancer chemotherapy: multicenter randomized trial. J Natl Cancer Inst 2013;105:1821–1832. [PubMed] [Google Scholar]
76. Friedenreich CM, Stone CR, Cheung WY, Hayes SC. Physical activity and mortality in cancer survivors: a systematic review and meta‐analysis. JNCI Cancer Spectr 2019;4. 10.1093/jncics/pkz080 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
77. Timmerman KL, Dhanani S, Glynn EL, Fry CS, Drummond MJ, Jennings K, et al. A moderate acute increase in physical activity enhances nutritive flow and the muscle protein anabolic response to mixed nutrient intake in older adults. Am J Clin Nutr 2012;95:1403–1412. [PMC free article] [PubMed] [Google Scholar]
78. Harber MP, Konopka AR, Undem MK, Hinkley JM, Minchev K, Kaminsky LA, et al. Aerobic exercise training induces skeletal muscle hypertrophy and age‐dependent adaptations in myofiber function in young and older men. J Appl Physiol 1985;2012:1495–1504. [PMC free article] [PubMed] [Google Scholar]
79. Létocart AJ, Mabesoone F, Charleux F, Couppé C, Svensson RB, Marin F, et al. Muscles adaptation to aging and training: architectural changes—a randomised trial. BMC Geriatr 2021;21:48. [PMC free article] [PubMed] [Google Scholar]
80. Cosaburi R. Principles of exercise training. CHEST 1992;101:263S–267S. [PubMed] [Google Scholar]
81. Starkie R, Ostrowski SR, Jauffred S, Febbraio M, Pedersen BK. Exercise and IL‐6 infusion inhibit endotoxin‐induced TNF‐α production in humans. FASEB J 2003;17:1–10. [PubMed] [Google Scholar]
82. Olsen RH, Krogh‐Madsen R, Thomsen C, Booth FW, Pedersen BK. Metabolic responses to reduced daily steps in healthy nonexercising men. JAMA 2008;299:1261–1263. [PubMed] [Google Scholar]
83. Rosenkilde M, Nordby P, Stallknecht B. Maintenance of improvements in fitness and fatness 1 year after a 3‐month lifestyle intervention in overweight men. Eur J Clin Nutr 2016;70:1212–1214. [PubMed] [Google Scholar]
84. Fedewa MV, Hathaway ED, Ward‐Ritacco CL. Effect of exercise training on C reactive protein: a systematic review and meta‐analysis of randomised and non‐randomised controlled trials. Br J Sports Med 2017;51:670–676. [PubMed] [Google Scholar]
85. Schmidt ME, Meynköhn A, Habermann N, Wiskemann J, Oelmann J, Hof H, et al. Resistance exercise and inflammation in breast cancer patients undergoing adjuvant radiation therapy: mediation analysis from a randomized, controlled intervention trial. Int J Radiat Oncol Biol Phys 2016;94:329–337. [PubMed] [Google Scholar]
86. Kleckner IR, Kamen C, Cole C, Fung C, Heckler CE, Guido JJ, et al. Effects of exercise on inflammation in patients receiving chemotherapy: a nationwide NCORP randomized clinical trial. Support Care Cancer 2019;27:4615–4625. [PMC free article] [PubMed] [Google Scholar]
87. Casuso RA, Huertas JR. Mitochondrial functionality in inflammatory pathology‐modulatory role of physical activity. Life (Basel) 2021;11:61. [PMC free article] [PubMed] [Google Scholar]
88. Brooks SV, Vasilaki A, Larkin LM, McArdle A, Jackson MJ. Repeated bouts of aerobic exercise lead to reductions in skeletal muscle free radical generation and nuclear factor κB activation. J Physiol 2008;586:3979–3990. [PMC free article] [PubMed] [Google Scholar]
89. Parise G, Phillips SM, Kaczor JJ, Tarnopolsky MA. Antioxidant enzyme activity is up‐regulated after unilateral resistance exercise training in older adults. Free Radic Biol Med 2005;39:289–295. [PubMed] [Google Scholar]
90. Kang X, Yang M, Shi Y, Xie M, Zhu M, Zheng X, et al. Interleukin‐15 facilitates muscle regeneration through modulation of fibro/adipogenic progenitors. Cell Commun Signal 2018;16:42. [PMC free article] [PubMed] [Google Scholar]
91. Adams GR, Zaldivar FP, Nance DM, Kodesh E, Radom‐Aizik S, Cooper DM. Exercise and leukocyte interchange among central circulation, lung, spleen, and muscle. Brain Behav Immun 2011;25:658–666. [PMC free article] [PubMed] [Google Scholar]
92. Batista ML, Rosa JC, Lopes RD, Lira FS, Martins E, Yamashita AS, et al. Exercise training changes IL‐10/TNF‐alpha ratio in the skeletal muscle of post‐MI rats. Cytokine 2010;49:102–108. [PubMed] [Google Scholar]
93. Pedersen L, Idorn M, Olofsson GH, Lauenborg B, Nookaew I, Hansen RH, et al. Voluntary running suppresses tumor growth through epinephrine‐ and IL‐6‐dependent NK cell mobilization and redistribution. Cell Metab 2016;23:554–562. [PubMed] [Google Scholar]
94. Garritson J, Krynski L, Haverbeck L, Haughian JM, Pullen NA, Hayward R. Physical activity delays accumulation of immunosuppressive myeloid‐derived suppressor cells. PLoS ONE 2020;15:e0234548. [PMC free article] [PubMed] [Google Scholar]
95. Pérez‐López A, McKendry J, Martin‐Rincon M, Morales‐Alamo D, Pérez‐Köhler B, Valadés D, et al. Skeletal muscle IL‐15/IL‐15Rα and myofibrillar protein synthesis after resistance exercise. Scand J Med Sci Sports 2018;28:116–125. [PubMed] [Google Scholar]
96. Conlon KC, Lugli E, Welles HC, Rosenberg SA, Fojo AT, Morris JC, et al. Redistribution, hyperproliferation, activation of natural killer cells and CD8 T cells, and cytokine production during first‐in‐human clinical trial of recombinant human interleukin‐15 in patients with cancer. J Clin Oncol 2015;33:74–82. [PMC free article] [PubMed] [Google Scholar]
97. Severinsen MCK, Schéele C, Pedersen BK. Exercise and browning of white adipose tissue—a translational perspective. Curr Opin Pharmacol 2020;52:18–24. [PubMed] [Google Scholar]
98. Duggal NA, Pollock RD, Lazarus NR, Harridge S, Lord JM. Major features of immunesenescence, including reduced thymic output, are ameliorated by high levels of physical activity in adulthood. Aging Cell 2018;17:e12750. [PMC free article] [PubMed] [Google Scholar]
99. Fischer CP. Interleukin‐6 in acute exercise and training: what is the biological relevance? Exerc Immunol Rev 2006;12:6–33. [PubMed] [Google Scholar]
100. Ostrowski K, Hermann C, Bangash A, Schjerling P, Nielsen JN, Pedersen BK. A trauma‐like elevation of plasma cytokines in humans in response to treadmill running. J Physiol 1998;513:889–894. [PMC free article] [PubMed] [Google Scholar]
101. Margeli A, Skenderi K, Tsironi M, Hantzi E, Matalas A‐L, Vrettou C, et al. Dramatic elevations of interleukin‐6 and acute‐phase reactants in athletes participating in the ultradistance foot race spartathlon: severe systemic inflammation and lipid and lipoprotein changes in protracted exercise. J Clin Endocrinol Metab 2005;90:3914–3918. [PubMed] [Google Scholar]
102. Ostrowski K, Schjerling P, Pedersen BK. Physical activity and plasma interleukin‐6 in humans—effect of intensity of exercise. Eur J Appl Physiol 2000;83:512–515. [PubMed] [Google Scholar]
103. Steensberg A, Keller C, Starkie RL, Osada T, Febbraio MA, Pedersen BK. IL‐6 and TNF‐α expression in, and release from, contracting human skeletal muscle. Am J Physiol Endocrinol Metab 2002;283:E1272–E1278. [PubMed] [Google Scholar]
104. Steensberg A, Fischer CP, Keller C, Møller K, Pedersen BK. IL‐6 enhances plasma IL‐1ra, IL‐10, and cortisol in humans. Am J Physiol‐Endocrinol Metab 2003;285:E433–E437. [PubMed] [Google Scholar]
105. Zanetti HR, da Cruz LG, Lourenço CLM, de Neves F, Silva‐Vergara ML, Mendes EL. Non‐linear resistance training reduces inflammatory biomarkers in persons living with HIV: A randomized controlled trial. Eur J Sport Sci 2016;16:1232–1239. [PubMed] [Google Scholar]
106. Weyh C, Krüger K, Strasser B. Physical activity and diet shape the immune system during aging. Nutrients 2020;12. [PMC free article] [PubMed] [Google Scholar]
107. Monteiro R, Azevedo I. Chronic inflammation in obesity and the metabolic syndrome. Mediat Inflamm 2010;2010:e289645. 10.1155/2010/289645 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
108. Kim JA, Kim SH, Kim IS, Yu DY, Kim SC, Lee SH, et al. Anti‐inflammatory effects of a mixture of lactic acid bacteria and sodium butyrate in atopic dermatitis murine model. J Med Food 2018;21(7):716–725. 10.1089/jmf.2017.4116 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
109. Bindels LB, Beck R, Schakman O, Martin JC, Backer FD, Sohet FM, et al. Restoring specific Lactobacilli levels decreases inflammation and muscle atrophy markers in an acute leukemia mouse model. PLoS ONE 2012;7:e37971. 10.1371/journal.pone.0037971 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
110. Bindels LB, Neyrinck AM, Claus SP, Le Roy CI, Grangette C, Pot B, et al. Synbiotic approach restores intestinal homeostasis and prolongs survival in leukaemic mice with cachexia. ISME J 2016;10:1456–1470. [PMC free article] [PubMed] [Google Scholar]
111. Wang J, Chen W‐D, Wang Y‐D. The relationship between gut microbiota and inflammatory diseases: the role of macrophages. Front Microbiol 2020;11:1065. [PMC free article] [PubMed] [Google Scholar]
112. Hakozaki T, Richard C, Elkrief A, Hosomi Y, Benlaïfaoui M, Mimpen I, et al. The gut microbiome associates with immune checkpoint inhibition outcomes in patients with advanced non‐small cell lung cancer. Cancer Immunol Res 2020;8:1243–1250. [PubMed] [Google Scholar]
113. Pötgens SA, Brossel H, Sboarina M, Catry E, Cani PD, Neyrinck AM, et al. Klebsiella oxytoca expands in cancer cachexia and acts as a gut pathobiont contributing to intestinal dysfunction. Sci Rep 2018;8:12321. [PMC free article] [PubMed] [Google Scholar]
114. Baruch EN, Youngster I, Ben‐Betzalel G, Ortenberg R, Lahat A, Katz L, et al. Fecal microbiota transplant promotes response in immunotherapy‐refractory melanoma patients. Science 2021;371:602–609. [PubMed] [Google Scholar]
115. Nay K, Jollet M, Goustard B, Baati N, Vernus B, Pontones M, et al. Gut bacteria are critical for optimal muscle function: a potential link with glucose homeostasis. Am J Physiol‐Endocrinol Metab 2019;317:E158–E171. [PubMed] [Google Scholar]
116. Estaki M, Pither J, Baumeister P, Little JP, Gill SK, Ghosh S, et al. Cardiorespiratory fitness as a predictor of intestinal microbial diversity and distinct metagenomic functions. Microbiome 2016;4:42. [PMC free article] [PubMed] [Google Scholar]
117. Clarke SF, Murphy EF, O'Sullivan O, Lucey AJ, Humphreys M, Hogan A, et al. Exercise and associated dietary extremes impact on gut microbial diversity. Gut 2014;63:1913–1920. [PubMed] [Google Scholar]
118. von Haehling S, Morley JE, Coats AJS, Anker SD. Ethical guidelines for publishing in the Journal of Cachexia, Sarcopenia and Muscle: update 2019. J Cachexia Sarcopenia Muscle 2019;10:1143–1145. [PMC free article] [PubMed] [Google Scholar]

Articles from Journal of Cachexia, Sarcopenia and Muscle are provided here courtesy of Wiley

-