Next Article in Journal
Development of Radiomic-Based Model to Predict Clinical Outcomes in Non-Small Cell Lung Cancer Patients Treated with Immunotherapy
Next Article in Special Issue
Is Acupuncture an Ideal Adjunctive Treatment for Cancer-Related Fatigue? Comment on Choi et al. Acupuncture for Managing Cancer-Related Fatigue in Breast Cancer Patients: A Systematic Review and Meta-Analysis. Cancers 2022, 14, 4419
Previous Article in Journal
Anoctamins and Calcium Signalling: An Obstacle to EGFR Targeted Therapy in Glioblastoma?
Previous Article in Special Issue
Acupuncture for Managing Cancer-Related Fatigue in Breast Cancer Patients: A Systematic Review and Meta-Analysis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Integrative Approaches to the Treatment of Cancer

1
NICM Health Research Institute, Western Sydney University, Westmead, NSW 2145, Australia
2
National Institute of Integrative Medicine, Hawthorn, VIC 3122, Australia
*
Author to whom correspondence should be addressed.
Cancers 2022, 14(23), 5933; https://doi.org/10.3390/cancers14235933
Submission received: 28 October 2022 / Revised: 21 November 2022 / Accepted: 22 November 2022 / Published: 30 November 2022
(This article belongs to the Special Issue Integrated Management of Cancer)

Abstract

:

Simple Summary

A significant proportion of people with cancer use forms of complementary medicine. Many factors contribute to cancer including some which are modifiable by the patient, such as stress, poor nutrition, vitamin D deficiency, poor sleep and lack of physical activity. This paper discusses why it is important that oncologists help cancer patients to address such factors, and why an integrative approach which combines evidence-based complementary medicines or therapies with orthodox treatment might lead to better outcomes for cancer patients.

Abstract

A significant proportion of cancer patients use forms of complementary medicine or therapies. An integrative approach to cancer management combines conventional medicine with evidence-based complementary medicines/therapies and lifestyle interventions, for the treatment and prevention of disease and the optimisation of health. Its basis is a holistic one; to treat the whole person, not just the disease. It makes use of adjunct technologies which may assist the clinician in diagnosis of early carcinogenesis and monitoring of treatment effectiveness. Many factors contribute to the development of cancer including some which are largely modifiable by the patient and which oncologists may be in a position to advise on, such as stress, poor nutrition, lack of physical activity, poor sleep, and Vitamin D deficiency. An integrative approach to addressing these factors may contribute to better overall health of the patient and better outcomes. Evidence-based complementary medicine approaches include the use of supplements, herbal medicine, various practices that reduce stress, and physical therapies. Individualised to the patient, these can also help address the symptoms and signs associated with cancer and its orthodox treatment.

1. Introduction

Cancer is a chronic, systemic illness with a variety of aetiological factors including genetic susceptibility, environmental, and epigenetic factors. Other associated factors include those that are largely modifiable by the individual, including stress, poor nutrition/diet, lack of physical activity, poor sleep, and vitamin D deficiency. Cancer and its orthodox treatment are associated with many symptoms and signs, many of which decrease the ability of patients to complete treatment, such as cancer-related pain, chemotherapy-induced peripheral neuropathy, oral mucositis, anxiety, depression, and poor sleep [1]. Many of these are not well managed with orthodox medicine approaches.
An integrative approach to the treatment of illness like cancer is one in which combines conventional medicine with evidence-based complementary medicines/therapies, nutritional medicine and lifestyle interventions for the treatment and prevention of disease and the optimisation of health. Its basis is a holistic one; to treat the whole person, not just the disease. It makes use of adjunct technologies which may assist the clinician in diagnosis of early carcinogenesis and monitoring of treatment effectiveness, such as circulating tumour cell tests [2,3,4,5]. Therapies that are adjunct to orthodox treatment can, importantly, help the cancer patient deal with many of the symptoms and signs associated with cancer and its orthodox treatment.
Indeed, there is much evidence to indicate that people with cancer and cancer survivors are using complementary medicines/therapies [6]. Proportions of cancer patients found to use complementary medicines/therapies range from around 40% in some studies to 84% in others [7,8,9,10,11].
An effective integrative approach facilitates a collaborative relationship between practitioner and patient, and importantly should empower the patient to be proactive in improving their health and well-being; that is, be involved in their self-care [12]. The notion of self-care was espoused in an Australian report, The State of Self Care in Australia which states that: “A healthy population is achieved through a functional relationship between active and informed individuals, health care services that empower and support people, and governments that invest in the capabilities of individual communities to look after their health” [13].
There is a growing interest in the field of oncology in relation to the integration of complementary therapies. Jentzsch and colleagues [14] concluded that in pancreatic ductal adenocarcinoma, an integrative treatment regimen combining first-line gemcitabine chemotherapy with two sub-groups of complementary medicines alternately in weekly cycles offers positive patient outcomes.
In this paper, we discuss key modifiable factors associated with cancer such as stress, nutrition, lack of physical activity, poor sleep and Vitamin D deficiency, including providing some of the evidence from an epidemiological research perspective of why these should be addressed. We will also briefly describe how these factors impact the pathomechanisms associated with cancer. An integrative approach that addresses these factors may contribute to better outcomes and better overall health of the patient. We examine some of the evidence that complementary medicines and approaches, including supplements, herbs and body therapies may be useful in an integrative approach to cancer treatment. Finally, we will offer our thoughts on how integrative oncology might be practised.

2. Addressing Modifiable Factors Associated with Cancer

Factors that can contribute to poor health are numerous; however, some key ones that are modifiable by individuals include stress, poor nutrition/diet, a lack of physical activity, poor sleep and inadequate levels of vitamin D, which have all been found to be associated with cancer [12]. These factors can contribute, at the very least, to a less than optimal quality of life. However, how do we relate these factors to what happens within the body when there is cancer?
The pathomechanism of cancer is complex. A seminal paper was published in 2000 on the “hallmarks of cancer” which set out an organizational framework of cellular properties uncovered during the transformation of normal cells to benign or malignant growths [15]. This was updated in 2011 with the inclusion of reprogramming of energy metabolism and evading immune destruction as additional hallmarks and the addition of two more enabling factors, tumor-promoting inflammation and genomic instability and mutation [16]. Then, in 2022 it was further updated to include phenotypic plasticity and disrupted differentiation as hallmark capabilities, and non-mutational epigenetic reprogramming and polymorphic microbiomes as enabling characteristics that can aid the acquisition of hallmark capabilities [17]. Kenny and colleagues [18], as well as many others (e.g., [16,17,19], draw attention to the importance of the tumor microenvironment and how it can influence the development and progression of tumors. The role of chronic low-grade inflammation in cancer is well established [17,20,21]. We can posit that factors such as stress and poor diet may contribute to chronic inflammation (discussed later) as part of Hanahan’s conceptual framework (the enabling factor of tumor-promoting inflammation).
The nervous system is also involved in cancer aetiology and pathogenesis [22,23]. Research indicates that tumour growth occurs in relation to the nervous system, indicating a functional role of nerves, neurons, neurites and neuroglia in tumorigenesis, discussed in depth by Baraldi and colleagues [22]. For example, tumour cells can release neurotrophins which stimulate adjacent neurites to grow into the tumour, and neurons can release neurotransmitters that initiate tumour cell migration [22]. Research also suggests sensory nerves may be able to regulate growth of tumours and metastasis by promoting or inhibiting immunosuppression [24]. The field of neuroimmunooncology is concerned with the interaction between the nervous system, immune system and cancer, and an effective approach to cancer may be one that targets the nervous system, and immune and/or genetic elements of the tumour micro/macro environments [22]. The concept of the neurobiology of cancer also recognizes the role of psychosocial factors in cancer [23].
Another conceptual framework, applied to nasopharygeal cancer (NPC) in a recent paper envisages NPC as a “spatiotemporal ‘unity of ecology and evolution’ disease: a multidimensional evolutionary adaptive pathological ecosystem” [25], describing how cancer tissues create a complex, spatially structured ecosystem of various cell types and essential stromal resources [25]. This framework could be extended to other forms of cancer to understand pathogenesis, as well as develop preventive strategies and therapeutic approaches (see [25]. Here, the concept of an ecosystem is applied at the tissue/molecular level to understand cancer pathogenesis, but this concept of an ecosystem could be extended outwards to a meta-level. That is, it could be used to describe the interdependence of humans and the environment in which we live and how, if this ecosystem becomes pathological, diseases like cancer can occur. The concept of environment can be broad, encompassing several dimensions such as the internal milieu within the body (i.e., internal environment, as described by Luo [25] and Hanahan [17]), the patient’s personal environment (e.g., immediate living and work environment), social/community environment and the greater environment (including climate, air quality, water quality, general standards of living, food supply and quality, and so on). All of these dimensions of “environment” are interdependent. The idea of a broader ecosystem is perhaps another way to understand how so many factors can contribute to cancer, and why the approach to its treatment should be multifactorial. This draws on one of the philosophies underpinning traditional Chinese medicine, as well as other systems of traditional medicine, considered to be of the “interdependence of man and environment” [26].
Whilst there are clearly many possible factors that can contribute to the development of cancer in an individual, we will focus on some key ones that are largely under the control of the patient, and for which doctors and oncologists may be able to offer assistance and/or advice.
In this section, we provide a rationale for why factors such as stress, poor sleep, poor diet, lack of physical activity and deficient Vitamin D levels should be addressed as part of an integrative approach to cancer management, drawing on just some of the evidence in the literature from epidemiological studies, as well as preclinical studies investigating mechanisms of action (though the latter will not be covered in depth due to the complexity of this area). Overall, we set out to stimulate thought on why such factors, those that patients can be empowered to address, ought to be considered by oncologists. In general, a healthy cancer patient is likely to do better than an unhealthy one.

2.1. Stress

The prevalence of psychological stress, anxiety and depression are major factors in cancer patients [27,28,29,30,31]. For example, the prevalence of depression may be anywhere from 25% to 66.7% [28,29,30], whilst the prevalence of anxiety has been found to be around 20% in one study [31].
Anxiety and depression are often comorbid [32] and both comorbid with other symptoms of cancer/cancer treatment, e.g., cancer-related fatigue [33], insomnia [34,35,36] and chronic pain [37,38]. Anxiety and depression can reduce quality of life and hinder cancer treatment, recovery and survival [39].
Some studies indicate that stressful life events can precede cancer [40,41] and that stress-related psychosocial factors are associated with higher cancer incidence and poorer survival [42]. Research suggests that depression may be associated with increased risk of cancer and may also predict cancer progression and decreased survival time [43,44,45,46].
There is evidence that chronic stress can induce tumorigenesis and promote cancer development [22,47], including influencing steps or pathways involved in metastatic spread [48]. Research in the field of psychoneuroimmunology has demonstrated ways in which the nervous system exerts complex effects on anti-cancer immunity [23]. Stress can impact on the immune system and endocrine systems, as well as the gut–brain axis (leading to abnormal gut flora) and these can impact on immunity and hormones (such as cortisol, growth hormone and prolactin) to promote tumour growth, as set out in Figure 1 [12]. Mravec [23] explains that cancer is also able to manipulate the nervous system. For example, it can induce new sympathetic nerves to grow into tumour tissue and cause the trans-differentiation of a sensory neuronal phenotype to adrenergic to utilise the stimulatory effect for adrenergic signalling, to promote cancer growth and metastasis.
Neurotransmitters released from nerves innervating tumor tissues can affect the growth of tumours as well as metastasis [23], and neurotransmitters involved in stress reactions have been found to impair the function of several subsets of leukocytes [22]. As pointed out by Baraldi and colleagues [22], immune suppression by the nervous system due to chronic stress, anxiety or depression, can facilitate tumour development.
From this we can begin to see a convergence of the fields of psychoneuroimmunology and neuroimmunooncology that may further elucidate how stress, anxiety and depression are involved in cancer.
Pathomechanisms by which chronic stress promotes cancer development are described in more detail in Dai et al. [47] and include the production of stress hormones (via activation of the hypothalamic-pituitary-adrenal axis and sympathetic nervous system), which then promote tumorigenesis and cancer development via several mechanisms. These include increasing p53 degradation, inducing DNA damage accumulation, increasing inflammation, suppressing the immune system and its surveillance functions, and acting on tumour cells and stromal cells within the tumour microenvironment, facilitating tumour growth, invasion and metastasis [47].
Loneliness and social isolation can impact on cancer, upregulating genes associated with gene transcripts involved with tumour progression and in high-risk patients, increasing intra-tumoural norepinephrine [49]. In a study of 9247 women with breast cancer, those who were socially isolated women (small networks) were 1.43 times more likely to have a breast cancer recurrence, 1.64 times more likely to die from breast cancer and 1.69 more likely to die of any cause than more socially integrated women [50]. Social support can affect cancer survival favourably [51,52,53]; for example, having at least one confidant reduced seven-year mortality by 39% in women with breast cancer [52].
A sensitive person will be more susceptible to depression and stress, compared to an insensitive type of person who may not even be aware of these factors.
Research indicates that many different approaches to stress reduction can be useful and improve quality of life in cancer patients. For example, practising transcendental meditation for 8 years was associated with a 49% decrease in rate of mortality from cancer [54]. Examples are set out in Table 1.
The Ornish Lifestyle Program is a lifestyle-driven approach used initially to control coronary artery disease (CAD) and other chronic diseases. It promotes lifestyle changes including a whole foods, plant-based diet low in fat, high in vegetables, fruits, wholegrains, legumes and soy products, and supplementation of Fish Oil, Vitamin E, Vitamin C and Selenium; smoking cessation; moderate exercise 30 min per day for 6 days; stress management techniques including yoga, meditation and breathing techniques, and weekly psychosocial support group meetings [68]. Studies in men with prostate cancer who had chosen not to undergo orthodox treatment found that those assigned to the lifestyle changes achieved significantly greater decreases in prostate-specific antigen (PSA) compared with those in the usual care control group [68] and had positive changes in gene expression over three months [69].

2.2. Disturbed Sleep

The prevalence of sleep disturbance in newly diagnosed or recently treated cancer patients is estimated to be 20–75% [70,71,72] and insomnia is found in 23–44% of patients 2–5 years after treatment for cancer [73,74]. Insomnia and sleep disturbances can lead to fatigue, mood disturbance, and contribute to immune suppression, affect quality of life, and may negatively impact on the disease course [75]. The National Cancer Institute [76] advises that up to 50% of cancer patients have sleep-related problems during treatment; these may be due to side effects of the treatment or pharmaceuticals, stress, long hospital stays and other factors.
Insomnia may be a risk factor for cancer, though as pointed out by Shi and colleagues [77], results of epidemiological studies have been somewhat equivocal as to whether there is a relationship between insomnia and cancer, with several suggesting a higher risk of cancer associated with insomnia [78,79,80,81] and others suggesting no association [82,83,84,85]. A meta-analysis conducted by Shi et al. [77] of these 8 studies found there was a modest 24% increased risk of cancer in those with insomnia (compared to those without insomnia). A recent review by Berisha and colleagues [86] indicates that chronic disruption of sleep/wake states prior to disease onset is associated with an increased risk of some cancers, e.g., breast, and that sleep disruption after cancer onset is often associated with poorer outcomes.
How disturbed sleep contributes to the development of cancer is complex. For example, mice experiments have shown that sleep characterized by frequent awakenings can speed the growth of cancer, increase tumor aggressiveness and depress immunity [87]. In sleep deprived mice, tumor associated macrophages (which can contribute to cancer progression by releasing different chemicals involved in tumour growth and invasion) were found to be more numerous and distributed closer to tumour capsules (compared with control mice) [87]. Sleep deprivation has been found to affect hundreds of genes related to circadian rhythms, metabolism, inflammation, immune response and stress [88]. As described by Shi et al. [77], other potential mechanisms of action proposed to explain how insomnia may contribute to cancer include dysregulation of melatonin [89] and circadian rhythm/chrono- disruptions (which can affect rhythmicity in neuroendocrine and immune parameters) [90], dysregulation of genes involved in tumour suppression [91], involvement of the oestrogen-signalling pathway [92], impaired immune function [93] and inflammation [94]. See Berisha et al. [86] and Shi et al. [77] for further discussion.
Approaches to improving sleep are many and include: general sleep hygiene guidelines, diet, exercise, stress reduction (e.g., relaxation/meditation/yoga), cognitive behavioural therapy, and supplements/herbs [12], including medicinal cannabis (discussed later). The US National Sleep Foundation’s 2013 Sleep in America Poll: Exercise and Sleep clearly shows beneficial effects of exercise on sleep, in particular, morning vigorous exercise [95].
Whilst it is beyond the scope of this paper to go into detail about different evidence-based complementary medicine approaches to each of the factors outlined above, readers are directed to any number of resources for further information, for example O’Brien and Sali [12] or Phelps [96]. In the following section, we will focus on examples of research into supplements and herbal medicines.

2.3. Diet and Nutrition

The link between diet and cancer has been revealed by the large variation in cancer rates between countries, as well as correlations with diet and observations of changes in cancer rates with migration [97,98]. Poor diet can lead to overweight and obesity, risk factors for cancer [99]: compared with those with a healthy weight, those with overweight or obesity have a greater risk of at least 13 types of cancer [100]. A 2019 study found that globally, excess body weight accounted for almost 4% of all cancers [101].
On the other hand, there are many studies which indicate that the adoption of particular diets such as the Mediterranean Diet are associated with reduced cancer incidence [102] and mortality [103].
Chronic, low-grade inflammation underpins many chronic illnesses such as cancer, as well as cardiovascular disease and type 2 diabetes, evidenced by elevations in levels of inflammatory biomarkers (e.g., C-reactive protein, interleukin 6 and 18, adhesion molecules (e.g., E selectin, intercellular adhesion molecule 1), vascular cell adhesion protein 1 and fibrinogen [104]. Diets high in sugar, saturated and trans-fatty acids and refined starches, and low in antioxidants, omega-3 polyunsaturated fatty acids (PUFAs) and fiber (e.g., from fruits, vegetables, whole grains) are proinflammatory and can activate the innate immune system, probably via increased production of proinflammatory cytokines and reduced production of anti-inflammatory cytokines [104]. The typical western diet has a ratio of the (more pro-inflammatory) omega 6 PUFAs to (anti-inflammatory) omega 3 PUFAs of between 10: and 30:1 [105,106,107], yet we evolved on a diet where this ratio was probably around 1:1 [105].
The Mediterranean Diet is characterized by high consumption of olive oil, fruits, vegetables, cereals (whole grains), legumes, seeds and nuts, moderate amounts of fish (a source of Omega 3 PUFAs), shellfish, white meat, eggs, fermented dairy products (e.g., cheese, yoghurt), and small amounts of red meat, processed meat and foods high in sugar, and includes the consumption of wine, especially red wine [104]. This diet has a high amount of omega 3 PUFAs (from fish and plants) and a low omega 6: omega 3 ratio of around 2:1 to 1:1 [104]. It has phytochemicals such as vitamin C, vitamin E, folate, carotenoids and polyphenols which have antioxidant and anti-inflammatory properties [104]. High dietary intake of antioxidants, including polyphenols associated with diets such as the Mediterranean Diet, may inhibit several cancer-related biological pathways, including reducing inflammation. Antioxidants play a role in cell differentiation and proliferation, and in synthesis and DNA repair by inhibiting production of carcinogenic chemicals endogenously, and reducing formation of adducts in DNA [104]. Omega 3 PUFAs can impact several of the cancer pathways including cell proliferation, cell survival (e.g., promoting apoptosis), inflammation, angiogenesis, metastasis and epigenetic abnormalities [108]. In animal models of breast cancer, diets high in extra virgin olive oil were found to induce different molecular changes in tumours, including in the activity of signalling molecules and gene expression, changes which induced lower proliferation, higher apoptosis and lower DNA damage compared with other diets [109].
Many studies have found that diets high in vegetables and fruit are protective against cancer and may play a role in prevention of disease recurrence [98,110,111,112,113,114,115,116]. Olive oil is also a key protective component compared to unstable vegetable oils used in the western diet. A systematic review found that high olive oil intake was associated with significant reduction in the risk of several cancers [117]. Yet, the Mediterranean Diet is not perfect; it includes preserved meats, wine and other foods which are not necessarily healthy. What might be protective may be the context of taking meals: meals are usually a social, family event. The social context of eating may be protective in itself [12]. It is very likely that the cultural aspects play a key role.
Diets that are high in meat and fat and low in dietary fibre are associated with greater risk of colorectal cancer (CRC) [118]. There is increasing evidence that CRC is occurring more frequently in younger people, with those consuming large amounts of deep-fried foods, refined foods, sugary drinks and desserts, and those on high fat diets and with low fibre and folate consumption being at higher risk [110].
Recent reviews of the role of dietary fibre reinforce the importance of these types of nutrients in the protection of cancer and other diseases [119,120]. Dietary fibre can influence metabolism in particular glucose and subsequent insulin, as well as have a major influence on microbiome [121,122]. The microbiota plays a role in bile acid metabolism, and epidemiological evidence supports the role of diet in modifying the composition and levels of bile acids, which in turn can modify the risk of colorectal cancer at the population level [118].

2.4. Inadequate Vitamin D

Research suggests a link between low Vitamin D levels and cancer. Systematic reviews have found associations between Vitamin D levels and risk of cancer, with higher levels being associated with lower risk [123,124,125,126]. In those with cancer, lower vitamin D levels are associated with greater cancer-related and all-cause mortality [127,128,129,130,131]. Inverse relationships are demonstrated between solar UVB and incidence and/or mortality rates for 22 types of cancer [132].
Vitamin D receptors are widely distributed in the body, including in immune response cells, supporting its role in homeostasis [133]. Vitamin D appears to play a role in protecting against cancer via several mechanisms, including inhibiting initiators of cellular angiogenesis (in various cancer cell lines), promoting antioxidant responses, inhibiting cell proliferation, stimulating DNA repair, suppressing metastasis and regulating autophagy [132,133]. Thus, a deficiency in Vitamin D could be relevant in the development of cancer [12].
Experimental studies indicate vitamin D has anti-neoplastic activity (apoptosis was induced by a metabolite of vitamin D in colorectal adenoma and carcinoma cells [134] and anti-proliferative activity [135]. Other preclinical studies (laboratory, animal) have found that vitamin D can inhibit carcinogenesis, slow tumour progression, inhibit cancer cell proliferation, promote apoptosis and is anti-inflammatory and anti-angiogenic [136].
Several studies have investigated the effect of vitamin D supplementation in cancer patients, with mixed results. For example, in a randomised controlled trial (RCT) of patients with digestive tract cancers, the AMATERASU trial in Japan, supplementation with 2000 IU vitamin D daily was not associated with any significant improvement in relapse-free survival at 5 years compared with placebo [137], though a post hoc age-adjusted analysis of the data actually showed a statistically significant benefit associated with supplementation (relapse-free survival HR, 0.66; 95% CI, 0.43–0.99) [136,137]. In a small study of 139 patients with advanced or metastatic colorectal cancer, high dose vitamin D (8000 IU daily for two weeks, and 4000 IU thereafter) was compared with standard dose vitamin D (400 IU daily) as an adjunct to standard chemotherapy. The high dose vitamin D was associated with a non-significant improvement in progression-free survival (13 months versus 11 months, p = 0.07) and significantly lower risk of progression-free survival or death (HR, 0.74; p  =  0.02); the latter effect was greater in those with lower BMI, prompting the researchers to opine that the results warranted larger scale studies [138].
A meta-analysis of RCTs found that over 2–7 years of supplementation with vitamin D had little effect on total cancer incidence (4 RCTs, n = 4333 participants, 400–1100 IU daily) but it was associated with significantly reduced total cancer mortality (3 RCTs, RR 0.88, 95% CI 0.78–0.98) [139]. In the large (n = 25,871) VITAL study in the US, vitamin D supplementation was not associated with a lower incidence of invasive cancer compared with placebo [140]; however, a secondary analysis indicated vitamin D supplementation was associated with a reduced risk of advanced (metastatic or fatal) cancer, with the strongest reduction in risk in those with normal weight [136].
Sunlight exposure is the best way to increase vitamin D, rather than via foods; however, when sunlight exposure is low, supplementation may be needed [12].

2.5. Inadequate Physical Activity

A meta-analysis of 17 prospective studies (total of 857,581 participants) found that sedentary behaviour significantly increased the risk of cancer by 20% [141]. Sedentary behaviour is associated with a higher risk of many cancers, for example endometrial [141,142], colorectal and its recurrence [141,142,143,144], breast cancer [141] and its recurrence [145], lung cancer [141], and prostate and ovarian cancer [142]. In addition, low levels of physical activity are associated with increased risk of all-cause and disease-specific mortality in cancer survivors [146]. Sedentary activity can be also associated with obesity, a risk factor for cancer [147], and there is evidence that weight loss can reduce risk of several cancers [148].
Positive news is that scientific evidence suggests that physical activity is protective against cancer [149,150,151]. In a meta-analysis which pooled the results of 12 prospective US and European cohorts (a total of 1.44 million subjects), in comparison with low levels of leisure-time physical activity, high levels of physical activity were significantly associated with lower risks of 13 cancers (10 of these associations remained statistically significant after taking body mass index into account) [151]. Exercise has been found to reduce risk factors for cancer such as obesity [152] and inflammation [153,154].
There are many benefits associated with exercising during and/or after cancer treatment [12], including helping protect against cancer-related fatigue [155] and improving health related outcomes in cancer survivors [156]. Physical activity after cancer diagnosis can reduce risk of all-cause death, as well as cancer-specific mortality [146,150,157,158]. Australian research found that in men with prostate cancer, exercise promoted the production of myokines (cytokines produced by muscle and secreted into the bloodstream) [159] which may be involved in exercise-induced tumour suppression [159,160]. Other benefits of exercise in cancer patients include improvements or preservation of muscle mass, strength and power, reduction in symptoms and side effects (including nausea, fatigue and pain), increased cardiorespiratory fitness, increased physical function, increased immune function, increased chemotherapy completion rates, reduced treatment-related side effects, improved curative effects of other treatments, better body image and self-esteem, decreased psychological and emotional distress, reduced depression and anxiety and shorter length of hospitalisation [161,162].
According to Friedenreich and colleagues [147], the molecular mechanisms by which sedentary lifestyles, lack of physical activity and obesity may contribute to cancer include effects on endogenous sex steroids and metabolic hormones, insulin sensitivity and chronic inflammation. Meanwhile, the mechanisms by which physical activity may reduce cancer risk are many and include decreasing systemic inflammation, hyperinsulinemia, insulin-like growth factor (IGF-I), sex hormones, pro-inflammatory leptin plus other cytokines associated with obesity, significantly increasing levels of (the anti-inflammatory) adipoleptin, improving immune function and the diversity and composition of the gut microbiome [149]. Other mechanisms include regulating cancer cell metabolism, regulating the immune environment, regulating growth factor secretion, targeting Akt and mTOR pathways, regulating skeletal muscle IL-6, and improving mitochondrial function which can inhibit cancer cell proliferation as well as apoptosis, though only moderate intensity exercise appears to exert significant influence on cancer cell proliferation and apoptosis [162]. During physical activity, contracting skeletal muscles release IL-6, a myokine, which exerts anti-inflammatory effects in other organs via an inflammatory (TNF-α) independent pathway, and the release of IL-6 induces increased anti-inflammatory interleukins IL-1ra and IL-10 [149]. Anti-inflammatory effects can occur via the reduction in visceral and body fat in addition to the anti-inflammatory mileu created through myokine release [149]. This helps explain how the anti-inflammatory effects of exercise are protective against systemic low-grade inflammation that underpins chronic illnesses such as cancer [149]. See Wang and Zhou [162] and Jurdana [149] for more information.

3. Complementary Medicines and Approaches in the Integrative Management of Cancer

Complementary medicine/treatment approaches may have beneficial effects in addressing many of the symptoms and signs associated with cancer and its orthodox treatment, as well as potentially addressing the pathomechanisms associated with cancer.
In this section, we describe just some of the evidence that complementary medicines or treatment approaches might be useful adjuncts in an integrative approach to cancer management.

3.1. Dietary Supplements

When a person does not have a sufficient variety of foods—particularly vegetables—in their diet, they can become deficient in vitamins and trace elements. Adequate micronutrients are essential for the proper functioning of the immune system and deficiency suppresses immunity [163]. Dietary supplements can help to ensure a person is as healthy as possible, where dietary sources of vitamins and minerals might be deficient [12]. For example, selenium has been primarily considered to have antioxidant, anti-inflammatory and anti-viral activity, but emerging evidence suggests a role in several of the pathways involved in cancer, including cell proliferation, migration, invasion and angiogenesis [164].
Another example of a type of supplement that can play an important role in an integrative approach to cancer is probiotics. The gut microbiome plays a critical role in the functioning of the immune system, influencing inflammation as well as the nervous system [165], and there is evidence that the bacterial microbiota plays a key role in carcinogenesis [166]. Stress can negatively impact the gut microbiome and chemotherapy and other drugs can damage it [12,167,168]. Chemotherapy can adversely have an impact on the gut microbiota, causing dysbiosis and altering its physiological and psychological functions [168]. Cancer treatments can alter the oral and gut microbiota and cause intestinal dysfunction, contributing to the pathogenesis of oral mucositis [167].
Other important supplements for cancer patients include: fish oil, vitamin C, co-enzyme Q10, magnesium, lycopene, vitamin E and vitamin B3 (this last one is in relation to skin cancer) [12,169].
There is evidence that antioxidants slow cancer progression and prevent its spread [170,171,172,173,174,175]. It is worth addressing the long-standing misperception that antioxidants might reduce the effectiveness of chemotherapy and radiation by reducing the potency of free radicals needed to kill cells [176]. Overall, research indicates that antioxidants in general (including low dose dietary forms and high dose IV forms) provide many benefits, do not reduce the efficacy of chemotherapy or radiation therapy, and can increase the effectiveness of conventional cancer therapeutic agents and decrease adverse effects [170,176,177,178,179,180]. For a concise explanation see Gonzales et al. [176]. A systematic review of 19 clinical trials investigating the use of antioxidants including glutathione (7), melatonin (4), Vitamin A (2), Vitamin C (1), Vitamin E (1), ellagic acid (1), N-acetylcysteine (1), and an antioxidant mixture (1), found that none of the studies reported evidence of significant decreases in chemotherapy efficacy due to antioxidant supplementation during chemotherapy [177]. Another systematic review also concluded that no trial reported a significant decrease in treatment efficacy with antioxidant use, and when antioxidants are included in a cancer patient’s therapeutic regime, there are several benefits, including reduced toxicity, improved treatment outcomes, increased survival times, increased tumour responses, and increased adherence to chemotherapy regimes [178].

3.2. Intravenous Supplements

The following supplements can be given at a much higher dose intravenously than orally: vitamin C, glutathione and A-Lipoic Acids. There is increasing evidence that high dose IV vitamin C has potential as a strong anti-cancer agent, with early phase clinical studies demonstrating efficacy in eradicating various types of cancer cells, as well as its safety [181].
The anti-cancer mechanisms of high doses of ascorbic acid (ascorbate) include pro-oxidant cytotoxic activity, inhibiting cell proliferation via inhibiting prostaglandins (2 series), inhibiting angiogenesis, anti-cancer epigenetic regulation, immune regulation, reversing epithelial-to-mesenchymal transition, inhibiting hypoxia and oncogenic kinase signalling and boosting the immune response [176,181,182]. High dose IV vitamin C is able to act synergistically with many standard chemotherapy drugs and mitigate their toxic side effects [181]. Adjunct IV Vitamin C administration for at least four weeks in breast cancer (stages IIa to IIIb) survivors undergoing chemotherapy or radiation therapy was associated with a significant reduction in side effects induced by the disease or the treatment, including nausea, loss of appetite, fatigue, depression, sleep disorders, dizziness and haemorrhagic diathesis compared to women without this adjunct treatment. There were no documented side effects of the IV vitamin C [183]. An important point in relation to surgery is that anaesthesia and surgery deplete vitamin C; humans cannot produce vitamin C, which is essential for brain function and wound healing [184]. Further clinical studies need to be carried out to evaluate the role of high dose IV Vitamin C for post-surgery treatment, including in cancer patients.
Vitamin C is relatively non-toxic at high levels. However, patients with glucose-6-phosphate deficiency may be at risk of developing haemolysis when given high doses of Vitamin C and hence patients need to be screened prior to high dose therapy.

3.3. Herbal Medicine

Many herbs and spices have been found to have anti-cancer properties in preclinical studies. Here are just a few examples.
Curcumin, a component of the well-known Asian spice turmeric (Curcuma longa), has anti-inflammatory, antiseptic, analgesic, antioxidant and anti-proliferative activity [185,186]. Curcumin has several anti-cancer actions via its effect on pathways involved in cell cycle regulation, apoptosis, mutagenesis, oncogene expression, tumorigenesis, and metastasis [186]. Curcumin can inhibit lung cell proliferation [187], inhibit prostate cancer cell growth via inhibition of androgen receptor pathways [188], inhibit prostate cancer bone metastasis [189], and inhibit epithelial mesenchymal transition and invasion induced by cancer-associated fibroblasts in prostate cells [190]. It can modulate many molecular targets, including transcription factors, growth factors and their receptors, cell adhesion molecules, enzymes, cytokines and genes involved in tumour growth, angiogenesis and metastasis [190,191]. Curcumin can modulate the growth of tumour cells through several cell signalling pathways, including cell proliferation, cell survival, caspas, tumour suppressor (p53, p21), death receptor, protein kinase and mitochondrial pathways [185]. Research in animals demonstrated its ability to inhibit tumour initiation and tumour promotion [185,192]. In humans, curcumin has also been found to be effective in treating depression and anxiety [193,194], which is relevant to cancer.
The herb cannabis sativa may be useful in the alleviation of several symptoms and signs associated with cancer and its orthodox treatment. These include cancer-related pain, cachexia, anxiety, depression, oral mucositis, chemotherapy-induced nausea and vomiting and sleep disorders [1]. Importantly, components of cannabis such as cannabidiol (CBD) may be protective against chemotherapy drug toxicity. CBD has demonstrated cardio-protective effects in animals against doxorubicin-induced cardiac injury and nephron-protective effects against cisplatin [195,196]. Mice studies found that CBD is protective against paclitaxel-induced neurotoxicity [197], though caution is advised with respect to immunotherapy drugs [198]. See O’Brien [1].
The Chinese herb Ganoderma lucidum (G. lucidum, Chinese herb Ling Zhi) has been shown to have anti-cancer properties. Preclinical research has demonstrated several anti-cancer mechanisms, including immunomodulation, inducing cell-cycle arrest and apotosis (several types of human tumour cells) and inhibiting cell adhesion, invasion and migration and angiogenic factors [199,200]. In animal experiments, a triterpene rich extract of G. lucidum was found to suppress prostate growth induced by testosterone [201], and one of its active constituents Ganoderol B was found to bind to the androgen receptor and inhibit 5α-reductase, suppress androgen-induced LNCaP cell growth, and downregulate Prostate-Specific Antigen (PSA) [202]. In a study of patients with advanced-stage cancers, supplementation with Ganopoly, the polysaccharides fractions extracted from G. lucidum, for 12 weeks resulted in significant enhancement of cellular immunity (elevated IL2, IL6 and interferon γ in 80% of patients [203]. A systematic review found that patients who had taken G. lucidum as an adjunct therapy with chemo/radiotherapy were more likely to respond positively compared to the orthodox treatment alone (RR 1.50; 95% CI 0.90–2.51, p = 0.02) [204]. Cao and colleagues [199] provide a comprehensive explanation of the research evidence for the anti-cancer actions of G. lucidum.
Chinese herbal medicine (CHM) typically uses combinations of herbs rather than single herbs. CHM may be a useful adjunct to orthodox treatment, enhancing the tumour response to chemotherapy and reducing toxicity [205]. It can also be useful in the treatment of many side effects of cancer and its orthodox treatment including, but not limited to the following: xerostomia following radiation therapy; nausea and vomiting (in particular associated with chemotherapy), other digestive problems (e.g., diarrhoea, constipation), anorexia, polyneuropathy, hot flushes, anxiety, depression, insomnia, pain and cancer-related fatigue [205,206,207,208].

3.4. Acupuncture

Evidence from randomised controlled trials suggests acupuncture is useful in the treatment of many symptoms and signs associated with cancer and its treatment, including chemotherapy-related nausea and vomiting, cancer fatigue, neutropenia, cancer-related pain and xerostomia [209,210,211,212,213,214]. For example, in a randomised controlled trial of 86 nasopharyngeal cancer patients, acupuncture was associated with positive effects on xerostomia as early as 3 weeks, with significantly greater saliva flow at week 7 and at the 6 months follow-up (p < 0.003). At the 6 months follow-up, 24% acupuncture group had xerostomia compared with 63% controls [210].

3.5. Massage

Scientific studies have demonstrated that oncology massage can reduce symptoms, including stress, pain, anxiety, depression, nausea and fatigue in those who have had surgery or chemotherapy for cancer [215]. Other benefits for cancer patients include improving coping, improvements in fatigue, pain and stress, enhanced quality of life, as well as improved immunity (lymphocyte count) [216,217,218].

4. Repurposing of Current Drugs and Their Influence on Metabolic Blockade

A potential source of new treatment options for cancer patients is the repurposing of existing non-cancer drugs. Off-label use of drugs is common in general practice and paediatrics where up to 10% of drugs are used off-label [219]. In particular, in oncology the practice of off-label use of drugs is even more common; in some studies up to 71% of adult cancer patients have been prescribed off-label drugs [220]. As the development of new cancer drugs is a challenging and costly endeavor, drug repurposing is regarded as an alternative to potentially accelerate this process.
There are several drugs which have been repurposed for the treatment of cancer. There is evidence that cancer cells share the same metabolic profile as trophoblastic cells, but different from adult somatic cells, e.g., use of glutamine, aerobic glycolysis [221]. Therefore, using off-label medications to block cancer metabolic pathways is a sound strategy in conjunction with standard care as its effect on non-cancer cells are limited and known to be safe in the doses used regularly for its on-label indications.
For example, studies have demonstrated evidence suggesting a potential role for metformin in cancer therapy. Preclinical studies have demonstrated several anti-cancer molecular mechanisms of metformin including mTOR inhibition, cytotoxic effects and immunomodulation. Epidemiologic data have demonstrated decreased cancer incidence and mortality in patients taking metformin. Several clinical trials, focused on evaluation of metformin as an anti-cancer agent are presently underway [222]. Metformin blunts the Warburg effect and consequently downregulates the growth of cancer stem cells. It has been suggested that metformin may well be used as a radiation sensitizer or an immunotherapy drug, in addition to a direct anti-proliferative agent for the treatment of cancer [223]. The use of metformin in diabetic patients diagnosed with cancer has been associated with a decrease in risk of cancer mortality [224].
Naltrexone (NTX) is an opioid antagonist traditionally used as a treatment for alcohol and opioid use disorders, but various studies have documented its involvement in cancer progression, when used as low dose treatment. Further evidence is necessary to demonstrate its efficacy and its mechanisms of action [225].
Melatonin has been used at high dosage, as an adjuvant in radiotherapy for radio-protection and radio-sensitization. The molecular mechanisms for radio-protection and radio-sensitizer effects of melatonin have been investigated. High dosages such as 60 mg, four times per day have been used to induce oncostasis and also reduce radiation side effects [226].
Dichloroacetate (DCA) works by turning on the natural cell suicide system, apoptosis, which is suppressed in cancerous cells, thus allowing their destruction. DCA does not poison the cells like cytotoxic chemotherapy drugs; it interferes with the utilisation of glucose by the cancer cell, leading to its starvation. It can improve the sensitivity to various drugs or radiotherapy, leading to apoptosis [227].
Sodium Phenylbutyrate (PB), a drug typically used in urea cycle disorders, is another useful drug that helps reduce serum levels of glutamine, which most cancers are dependent on without affecting normal cells. Cancer cells are well known to be addicted to glutamine as its major fuel source apart from glucose [228]. Lowering serum levels with phenylbutyrate helps to deprive one of their main sources of energy. PB also helps to induce cell differentiation, which makes for a less aggressive phenotype, a particular useful strategy in cancer therapy [229]. It is also a histone deacetylase inhibitor, which helps to reduce gene expression in cancer cells [230].
Several other drugs have been explored because of their repurposing potential, including Sodium Phenylbutyrate; Atorvastatin; Dipyridamole; Doxycycline and Mebendazole.
However, although repurposing drugs is an appealing strategy, this is no substitute for randomized clinical trials to determine efficacy and the populations most likely to benefit from an intervention.

5. Other Therapies for Cancer Treatment

There are other therapies which may be useful in the treatment of cancer. The National Institute of Integrative Medicine in Australia has been investigating the role of Photodynamic Therapy for the treatment of prostate cancer with promising preliminary results. The laser is delivered to the prostate trans-rectal and trans-urethral (see Meade et al. [231]). Hyperthermia and hyperbaric therapy are increasingly being used in the treatment of cancer, but due to limited capacity within this paper, details are not included.

6. Mitigating Risk in an Integrative Approach to Cancer Treatment

There are naturally concerns about whether any nutritional supplements and herbal medicines could adversely interact or interfere with orthodox cancer treatment. The potential for adverse interactions between certain drugs and certain forms of complementary medicines is well documented in the literature [232,233,234,235]. On the other hand, many complementary medicines have benefits in combination with orthodox cancer treatment (e.g., [1,137]. For example, the use of a common Chinese herbal formula called Jia Wei Xiao Yao San with Tamoxifam reduced the risk of subsequent endometrial cancer in female breast cancer patients [236].
There are drug-supplement/herb databases that set out the scientific evidence in relation to potential interactions, such as the IMGateway, an online portal for healthcare practitioners which is being adapted for consumer use [6]. Use of such evidence-based portals by practitioners can reduce potential for adverse interactions and provide a level of confidence for both patient and practitioner.

7. How to Practise Integrative Oncology

It is not possible to be an expert in oncology as well as nutritional medicine, stress reduction techniques, herbal medicine and so on. However, doctors can inquire about those factors that can impact on cancer patients (covered earlier in the paper) and facilitate referral to other healthcare practitioners capable of assisting. A team approach that integrates input from clinicians with different types of expertise is ideal, and often the general practitioner has an important role to play here. Remember, research indicates that a significant proportion of patients (42% in one US study) do not speak to their doctor about their complementary medicine use, often fearing disapproval [237]. Those patients who want to use complementary medicine approaches are likely to do so with or without your approval. Therefore, it is important to keep an open mind, and read and critique the scientific evidence available on complementary medicine approaches—there is much more research evidence than many think.
Deciding which complementary medicines or approaches to integrate with standard therapy can be done systematically and rationally, as exemplified by Jentzsch and colleagues [14]. In their paper, they set out an evidence-based approach to choosing which complementary medicines/therapies/approaches should be combined with gemcitabine in the treatment of patients with PDAC. They firstly divided the various complementary measures into three groups: dietary factors, nutraceutical agents and lifestyle. They then looked at the available evidence in relation to specific dietary and nutraceutical agents, considering clinical trials, meta-analyses, in vivo tests and in vitro studies. From this they were able to identify 9 agents: 6 dietary (Vitamins A, C, D and E, genistein and curcumin) and 3 nutraceutical compounds (propolis, triptolide and cannabidiol) that they deemed acceptable due to the available evidence base, for integration with gemcitabine chemotherapy. They propose an integrative treatment regimen combining gemcitabine chemotherapy with the two sub-groups of complementary agents alternately in weekly cycles could then be applied, with the ability to modify this protocol for poor responders or “super responders”. The judicious combining of particular complementary agents took into consideration their main mechanisms of action in relation to particular hallmarks of cancer [14]. This methodology could be readily adapted to other types of cancer and is an example of a systematic, rational approach to considering what types of complementary medicines might be safely combined with standard oncology treatment, taking into consideration safety issues such as potential interactions.

8. Conclusions

An integrative approach to cancer treatment uses the best of evidence-based conventional and complementary medicine approaches. It empowers the patient to be proactive in improving their overall health and addressing many of the factors associated with cancer, as well as symptoms and signs associated with cancer and its orthodox treatment. Helping the cancer patient address factors that are largely within their control such as stress, poor nutrition, poor sleep, vitamin D deficiency and lack of physical exercise may result in a better quality of life and better outcomes. There is much scientific research in the literature—preclinical and clinical—indicating that many forms of complementary medicine are effective in alleviating many of the symptoms/signs associated with cancer and its treatment, and may potentially also address the pathomechanisms underpinning various cancers. Cancer patients are using complementary medicines. By becoming better informed, doctors are in a more nuanced position to make recommendations to patients, as well as refer to a wider network of healthcare professionals as part of a team approach to cancer care.

Author Contributions

This paper was conceptualized by A.S. and K.O. K.O. was responsible for writing the article, with T.B. contributing the section on repurposed drugs, K.R. contributing information on circulating tumour cells and providing feedback on early drafts and A.S. also providing additional references and feedback on content. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. O’Brien, K. Cannabidiol (CBD) in Cancer Management. Cancers 2022, 14, 885. [Google Scholar] [CrossRef]
  2. Cristofanilli, M.; Budd, G.T.; Ellis, M.J.; Stopeck, A.; Matera, J.; Miller, M.C.; Reuben, J.M.; Doyle, G.V.; Allard, W.J.; Terstappen, W.M.M.; et al. Circulating tumor cells, disease progression, and survival in metastatic breast cancer. N. Engl. J. Med. 2004, 351, 781–791. [Google Scholar] [CrossRef] [Green Version]
  3. Rarecells Diagnostics, France. Available online: https://www.rarecells.com/scientific-publications (accessed on 16 November 2022).
  4. Ried, K.; Eng, P.; Sali, A. Screening for Circulating Tumour Cells allows early detection of cancer and monitoring of treatment effectiveness: An observational study. Asian Pac. J. Cancer Prev. 2017, 18, 2275–2285. [Google Scholar] [CrossRef] [Green Version]
  5. Ried, K.; Tamanna, T.; Matthews, S.; Eng, P.; Sali, A. New Screening Test Improves Detection of Prostate Cancer Using Circulating Tumor Cells and Prostate-Specific Markers. Front. Oncol. 2020, 10, 582. [Google Scholar] [CrossRef] [Green Version]
  6. O’Brien, K.; Moore, A.; Percival-Smith, S.; Venkatraman, S.; Grubacevic, V.; Scoble, J.; Gilham, L.; Greenway, T.; Coghill, K.; Wale, J.; et al. An investigation into the usability of a drug-complementary medicines interactions database in a consumer group of women with breast cancer. Eur. J. Integr. Med. 2020, 33, 101004. [Google Scholar] [CrossRef]
  7. Ernst, E.; Cassileth, B.R. The prevalence of complementary/alternative medicine in cancer: A systematic review. Cancer 1998, 83, 777–782. [Google Scholar] [CrossRef]
  8. Kang, D.H.; McArdle, R.; Suh, Y. Changes in complementary and alternative medi-cine use across cancer treatment and relationship to stress, mood, and quality of life. J. Altern. Complement. Med. 2014, 20, 853–859. [Google Scholar] [CrossRef] [Green Version]
  9. Morris, K.T.; Johnson, N.; Homer, L.; Walts, D. A comparison of complementary therapy use between breast cancer patients and patients with other primary tumor sites. Am. J. Surg. 2000, 179, 407–411. [Google Scholar] [CrossRef]
  10. Richardson, M.A.; Sanders, T.; Palmer, J.L.; Greisinger, A.; Singletary, S.E. Complementary/alternative medicine use in a comprehensive cancer center and the implications for oncology. J. Clin. Oncol. 2000, 18, 2505–2514. [Google Scholar] [CrossRef]
  11. Wyatt, G.; Sikorskii, A.; Wills, C.E.; Su, H. Complementary and Alternative Medicine Use, Spending, and Quality of Life in Early Stage Breast Cancer. Nurs. Res. 2010, 59, 58–66. [Google Scholar] [CrossRef]
  12. O’Brien, K.; Sali, A. A Clinician’s Guide to Integrative Oncology. What You Should Be Talking about with Cancer Patients and Why; Springer: Cham, Switzerland, 2017. [Google Scholar]
  13. Australian Health Policy Collaboration. The State of Self Care in Australia. Available online: https://www.chpaustralia.com.au/Self-Care/Research (accessed on 10 October 2022).
  14. Jentzsch, V.; Davis, J.A.A.; Djamgoz, M.B. Pancreatic cancer (PDAC): Introduction of evidence-based complementary measures into integrative clinical management. Cancers 2020, 12, 3096. [Google Scholar] [CrossRef] [PubMed]
  15. Hanahan, D.; Weinberg, R.A. The hallmarks of cancer. Cell 2000, 100, 57–70. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Hanahan, D. Hallmarks of Cancer: New Dimensions. Cancer Discov. 2022, 12, 31–46. [Google Scholar] [CrossRef] [PubMed]
  18. Kenny, P.A.; Nelson, C.M.; Bissell, M.J. The ecology of tumors: By perturbing the microenvironment, wounds and infection may be key to tumor development. Scientist 2006, 20, 30. [Google Scholar] [PubMed]
  19. Anderson, M.N.; Simon, M.C. The tumor microenvironment. Curr. Biol. 2020, 30, R921–R925. [Google Scholar] [CrossRef]
  20. Kim, Y.S.; Young, M.R.; Bobe, G.; Colburn, N.H.; Milner, J.A. Bioactive food components, inflammatory targets, and cancer prevention. Cancer Prev. Res. Phila. 2009, 2, 200–208. [Google Scholar] [CrossRef] [Green Version]
  21. Schäfer, M.; Werner, S. Cancer as an overhealing wound: An old hypothesis revisited. Nat. Rev. Mol. Cell Biol. 2008, 9, 628–638. [Google Scholar] [CrossRef]
  22. Baraldi, J.H.; Martyn, G.V.; Shurin, G.V.; Shurin, M.R. Tumor innervation: History, methodologies, and significance. Cancers 2022, 14, 1979. [Google Scholar] [CrossRef]
  23. Mravec, B. Neurobiology of cancer: Definition, historical overview, and clinical implications. Cancer Med. 2022, 11, 903–921. [Google Scholar] [CrossRef]
  24. Scheff, N.N.; Saloman, J.L. Neuroimmunology of cancer and associated symptomology. Immunol. Cell Biol. 2021, 99, 949–961. [Google Scholar] [CrossRef] [PubMed]
  25. Luo, W. Nasopharyngeal carcinoma ecology theory: Cancer a multidimensional spatiotermporal “unity of ecology and evolution” pathological ecosystem. Preprints 2022, 2022100226. [Google Scholar]
  26. O’Brien, K.A.; Xue, C.C. The theoretical framework of Chinese medicine. In A Comprehensive Guide to Chinese Medicine, 2nd ed.; Leung, P.-C., Xue, C.C., Cheng, Y.-C., Eds.; World Scientific Press: Hackensack, NJ, USA, 2016. [Google Scholar]
  27. Ayalew, M.; Deribe, B.; Duko, B.; Geleta, D.; Bogale, N.; Gemechu, L.; Gebretsadik, A.; Bedaso, A. Prevalence of depression and anxiety symptoms and their determinant factors among patients with cancer in southern Ethiopia: A cross-sectional study. BMJ Open 2022, 12, e051317. [Google Scholar] [CrossRef] [PubMed]
  28. Henriksson, M.M.; Isometsä, E.T.; Hietanen, P.S.; Aro, H.M.; Lonnqvist, J.K. Mental disorders in cancer suicides. J. Affect. Disord. 1995, 36, 11–20. [Google Scholar] [CrossRef]
  29. Hong, J.S.; Tian, J. Prevalence of anxiety and depression and their risk factors in Chinese cancer patients. Support. Care Cancer 2013, 22, 453–459. [Google Scholar] [CrossRef]
  30. Lloyd-Williams, M. Depression- the hidden symptom in advanced cancer. J. Royal. Soc. Med. 2003, 96, 577–581. [Google Scholar]
  31. Naser, A.Y.; Hameed, A.N.; Mustafa, N.; Alwafi, H.; Dahmash, E.Z.; Alyami, H.S.; Khalil, H. Depression and Anxiety in Patients With Cancer: A Cross-Sectional Study. Front. Psychol. 2021, 12, 585534. [Google Scholar] [CrossRef]
  32. Gold, M.; Dunn, L.B.; Phoenix, B.; Paul, S.M.; Hamolsky, D.; Levine, J.D.; Miaskowski, C. Co-occurrence of anxiety and depressive symptoms following breast cancer surgery and its impact on quality of life. Eur. J. Oncol. Nurs. 2016, 20, 97–105. [Google Scholar] [CrossRef] [Green Version]
  33. Ho, R.T.H.; Kwan, T.T.C.; Cheung, I.K.M.; Chan, C.K.P.; Lo, P.H.Y.; Yip, P.S.F.; Luk, M.-Y.; Chan, C.L.W. Association of Fatigue with Perceived Stress in Chinese Women with Early Stage Breast Cancer Awaiting Adjuvant Radiotherapy. Stress Health 2015, 31, 214–221. [Google Scholar] [CrossRef]
  34. Baglioni, C.; Battagliese, G.; Feige, B.; Spiegelhalder, K.; Nissen, C.; Voderholzer, U.; Lombardo, C.; Riemann, D. Insomnia as a predictor of depression: A meta-analytic evaluation of longitudinal epidemiological studies. J. Affect. Disord. 2011, 135, 10–19. [Google Scholar] [CrossRef]
  35. Johnson, E.O.; Roth, T.; Breslau, N. The association of insomnia with anxiety orders and depression: Exploration of the direction of risk. J. Psychiatr. Res. 2006, 40, 700–708. [Google Scholar] [CrossRef] [PubMed]
  36. Taylor, D.J.; Lichstein, K.L.; Durrence, H.H.; Reidel, B.W.; Bush, A.J. Epidemiology of Insomnia, Depression, and Anxiety. Sleep 2005, 28, 1457–1464. [Google Scholar] [CrossRef] [PubMed]
  37. Arora, A.; Saini, S.; Nautiyal, V.; Verma, S.; Gupta, M.; Kalra, B.; Ahmad, M. Cancer pain, anxiety, and depression in admitted patients at a tertiary care hospital: A prospective observational study. Indian J. Palliat. Care 2019, 25, 562–566. [Google Scholar] [CrossRef] [PubMed]
  38. Li, W.; Xu, Y.; Luo, X.; Wen, Y.; Ding, K.; Xu, W.; Garg, S.; Yang, Y.; Sun, H. Alleviating excessive worries improves co-occurring depression and pain in adolescent and young adult cancer patients: A network approach. Neuropsychiatr. Dis. Treat. 2022, 18, 1843–1854. [Google Scholar] [CrossRef] [PubMed]
  39. Niedzwiedz, C.L.; Knifton, L.; Robb, K.A.; Katikireddi, S.V.; Smith, D.J. Depression and anxiety among people living with and beyond cancer: A growing clinical and research priority. BMC Cancer 2019, 19, 1–8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Fagundes, C.P.; Glaser, R.; Malarkey, W.B.; Kiecolt-Glaser, J.K. Childhood adversity and herpesvirus latency in breast cancer survivors. Health Psychol. 2013, 32, 337–344. [Google Scholar] [CrossRef] [Green Version]
  41. Lillberg, K.; Verkasalo, P.K.; Kaprio, J.; Teppo, L.; Helenius, H.; Koskenvuo, M. Stressful Life Events and Risk of Breast Cancer in 10,808 Women: A Cohort Study. Am. J. Epidemiol. 2003, 157, 415–423. [Google Scholar] [CrossRef] [Green Version]
  42. Chida, Y.; Hamer, M.; Wardle, J.; Steptoe, A. Do stress-related psychosocial factors contribute to cancer incidence and survival? Nat. Rev. Clin. Oncol. 2008, 5, 466–475. [Google Scholar] [CrossRef]
  43. Brown, K.W.; Levy, A.R.; Rosberger, Z.; Edgar, L. Psychological distress and cancer survival: A follow-up 10 years after diagnosis. Psychosom. Med. 2003, 65, 636–643. [Google Scholar] [CrossRef]
  44. Oerlemans, M.E.; Akker, M.V.D.; Schuurman, A.G.; Kellen, E.; Buntinx, F. A meta-analysis on depression and subsequent cancer risk. Clin. Pract. Epidemiol. Ment. Health 2007, 3, 29. [Google Scholar] [CrossRef] [Green Version]
  45. Penninx, B.W.J.H.; Guralnik, J.M.; Havlik, R.J.; Pahor, M.; Ferrucci, L.; Cerhan, J.; Wallace, R.B. Chronically Depressed Mood and Cancer Risk in Older Persons. JNCI J. Natl. Cancer Inst. 1998, 90, 1888–1893. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Spiegel, D.; Giese-Davis, J. Depression and cancer: Mechanisms and disease progression. Biol. Psychiatr. 2003, 54, 269–282. [Google Scholar] [CrossRef] [PubMed]
  47. Dai, S.; Mo, Y.; Wang, Y.; Xiang, B.; Liao, Q.; Zhou, M.; Li, X.; Li, Y.; Xiong, W.; Li, G.; et al. Chronic Stress Promotes Cancer Development. Front. Oncol. 2020, 10, 01492. [Google Scholar] [CrossRef] [PubMed]
  48. Lutgendorf, S.K.; Sood, A.K. Biobehavioural factors and cancer progression: Physiological pathways and mechanisms. Psychosom. Med. 2011, 73, 724–730. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Lutgendorf, S.K.; DeGeest, K.; Sung, C.Y.; Arevalo, J.M.; Penedo, F.; Lucci, J.; Goodheart, M.; Lubaroff, D.; Farley, D.M.; Sood, A.K.; et al. Depression, social support, and beta-adrenergic transcription control in human ovarian cancer. Brain, Behav. Immun. 2009, 23, 176–183. [Google Scholar] [CrossRef] [Green Version]
  50. Kroenke, C.H.; Michael, Y.L.; Poole, E.M.; Kwan, M.L.; Nechuta, S.; Leas, E.; Caan, B.J.; Pierce, J.; Shu, X.-O.; Zheng, Y.; et al. Postdiagnosis social networks and breast cancer mortality in the After Breast Cancer Pooling Project. Cancer 2016, 123, 1228–1237. [Google Scholar] [CrossRef] [Green Version]
  51. Fawzy, F.I.; Canada, A.L.; Fawzy, N.W. Malignant melanoma: Effects of a brief, structured psychiatric intervention on survival and recurrence at 10-year follow-up. Arch. Gen. Psychiatr. 2003, 60, 100–103. [Google Scholar] [CrossRef] [Green Version]
  52. Maunsell, E.; Brisson, J.; Deschěnes, L. Social support and survival among women with breast cancer. Cancer 1995, 76, 631–637. [Google Scholar] [CrossRef]
  53. Spiegel, K.; Bloom, J.R.; Kraemer, H.C.; Gotthiel, E. Effect of psycho-social treatment on survival of patients with metastatic breast cancer. Lancet 1989, 2, 888–891. [Google Scholar] [CrossRef]
  54. Schneider, R.H.; Alexander, C.N.; Staggers, F.; Rainforth, M.; Salerno, J.W.; Hartz, A.; Arndt, S.; Barnes, V.A.; Nidich, S.I. Long-Term Effects of Stress Reduction on Mortality in Persons ≥55 Years of Age With Systemic Hypertension. Am. J. Cardiol. 2005, 95, 1060–1064. [Google Scholar] [CrossRef] [Green Version]
  55. Meares, A. Regression of cancer after intensive meditation. Med. J. Aust. 1976, 2, 184. [Google Scholar] [CrossRef] [PubMed]
  56. Orme-Johnson, D. Medical care utilisation and the transcendental meditation program. Psychosom. Med. 1987, 49, 493–507. [Google Scholar] [CrossRef] [PubMed]
  57. Zeng, Y.; Luo, T.; Xie, H.; Huang, M.; Cheng, A.S. Health benefits of qigong or tai chi for cancer patients: A systematic review and meta-analyses. Complement. Ther. Med. 2014, 22, 173–186. [Google Scholar] [CrossRef] [PubMed]
  58. Fleishman, S.B.; Homel, P.; Chen, M.R.; Rosenwald, V.; Abolencia, V.; Gerber, J.; Nadesan, S. Beneficial effects of animal-assisted visits on quality of life during multimodal radiation-chemotherapy regimes. J. Community Support. Oncol. 2015, 13, 22–26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Orlandi, M.; Trangeled, K.; Mambrini, A.; Tagliani, M.; Ferrarini, A.; Zanetti, L.; Tartarini, R.; Pacetti, P.; Cantore, M. Pet therapy effects on oncological day hospital patients undergoing chemotherapy treatment. Anticancer. Res. 2008, 27, 4301–4303. [Google Scholar]
  60. Bar-Sela, G.; Atid, L.; Danos, S.; Gabay, N.; Epelbaum, R. Art therapy improved depression and influenced fatigue levels in cancer patients on chemotherapy. Psychooncology 2007, 16, 980–984. [Google Scholar] [CrossRef]
  61. Lawson, L.M.; Williams, P.; Glennon, C.; Carithers, K.; Schnabel, E.; Andrejack, A.; Wright, N. Effect of art making on cancer-related symptoms of blood and marrow transplantation recipients. Oncol. Nurs. Forum. 2012, 39, E353–E360. [Google Scholar] [CrossRef] [Green Version]
  62. Nainis, N.; Paice, J.A.; Ratner, J.; Wirth, J.H.; Lai, J.; Shott, S. Relieving Symptoms in Cancer: Innovative Use of Art Therapy. J. Pain Symptom Manag. 2006, 31, 162–169. [Google Scholar] [CrossRef]
  63. Stuckey, H.L.; Nobel, J. The Connection Between Art, Healing, and Public Health: A Review of Current Literature. Am. J. Public Health 2010, 100, 254–263. [Google Scholar] [CrossRef]
  64. Thyme, K.E.; Sundin, E.C.; Wiberg, B.; Öster, I.; Åström, S.; Lindh, J. Individual brief art therapy can be helpful for women with breast cancer: A randomized controlled clinical study. Palliat. Support. Care 2009, 7, 87–95. [Google Scholar] [CrossRef]
  65. Bradt, J.; Dileo, C.; Magill, L.; Teague, A. Music interventions for improving psychological and physical outcomes in cancer patients. Cochrane Database Syst. Rev. 2016, 15, CD006911. [Google Scholar] [CrossRef]
  66. Bradt, J.; Dileo, C.; Myers-Coffman, K.; Biondo, J. Music interventions for improving psychological and physical outcomes in cancer patients. Cochrane Database Syst. Rev. 2021, 10, CD006911. [Google Scholar] [PubMed]
  67. Morgan, N.P.; Graves, K.D.; Poggi, E.A.; Cheson, B.D. Implementing an Expressive Writing Study in a Cancer Clinic. Oncologist 2008, 13, 196–204. [Google Scholar] [CrossRef]
  68. Ornish, D.; Weidner, G.; Fair, W.R.; Marlin, R.; Pettengill, E.B.; Raisin, C.J.; Dunn-Emke, S.; Crutchfield, L.; Jacobs, F.N.; Barnard, R.J.; et al. Intensive lifestyle changes may affect the progression of prostate cancer. J. Urol. 2005, 174, 1065–1070. [Google Scholar] [CrossRef]
  69. Ornish, D.; Magbanua, M.J.M.; Weidner, G.; Weinberg, V.; Kemp, C.; Green, C.; Mattie, M.D.; Marlin, R.; Simko, J.; Shinohara, K.; et al. Changes in prostate gene expression in men undergoing an intensive nutrition and lifestyle intervention. Proc. Natl. Acad. Sci. USA 2008, 105, 8369–8374. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Fiorentino, L.; Ancoli-Israel, S. Sleep dysfunction in patients with cancer. Curr. Treat Options Neurol. 2007, 9, 337–346. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Fiorentino, L.; Rissling, M.; Liu, L.; Ancoli-Israel, S. The symptom cluster of sleep, fatigue and depressive symptoms in breast cancer patients: Severity of the problem and treatment options. Drug Discov. Today Dis. Model. 2012, 8, 167–173. [Google Scholar] [CrossRef] [Green Version]
  72. Harrold, E.C.; Idris, A.F.; Keegan, N.M.; Corrigan, L.; Teo, M.Y.; O’Donnell, M.; Lim, S.T.; Duff, E.; O’Donnell, D.M.; Kennedy, M.J.; et al. Prevalence of Insomnia in an Oncology Patient Population: An Irish Tertiary Referral Center Experience. J. Natl. Compr. Cancer Netw. 2020, 18, 1623–1630. [Google Scholar] [CrossRef]
  73. Engstrom, C.A.; Strohl, R.A.; Rose, L.; Lewandowski, L.; Stefanek, M.E. Sleep alterations in cancer patients. Cancer Nurs. 1999, 22, 143–148. [Google Scholar] [CrossRef]
  74. Vachani, C. Sleep Problems (Insomnia) in the Cancer Patient. 2016. OncoLink. Available online: https://www.oncolink.org/support/side-effects/insomnia/sleep-problems-insomnia-in-the-cancer-patient (accessed on 4 January 2017).
  75. O’Donnell, J.F. Insomnia in cancer patients. Clin. Cornerstone 2004, 6, S6–S14. [Google Scholar] [CrossRef]
  76. National Cancer Institute. Sleep Problems in People with Cancer. Reviewed 12 August 2021. Available online: https://www.cancer.gov/about-cancer/treatment/side-effects/sleep-problems (accessed on 19 November 2022).
  77. Shi, T.; Min, M.; Sun, C.; Zhang, Y.; Liang, M.; Sun, Y. Does insomnia predict a high risk of cancer? A systematic review and meta-analysis of cohort studies. J. Sleep Res. 2019, 29, e12876. [Google Scholar] [CrossRef] [PubMed]
  78. Chiu, H.Y.; Huang, C.J.; Fan, Y.C.; Tsai, P.S. Insomnia but not hypnotics use associates with the risk of breast cancer: A popula- tion-based matched cohort study. J. Womens Health 2018, 27, 1250–1256. [Google Scholar] [CrossRef] [PubMed]
  79. Luo, J.; Sands, M.; Wactawski-Wende, J.; Song, Y.; Margolis, K.L. Sleep disturbance and incidence of thyroid cancer in postmenopausal women the Women’s Health Initiative. Am. J. Epidemiol. 2013, 177, 42–49. [Google Scholar] [CrossRef]
  80. Sen, A.; Opdahl, S.; Strand, L.B.; Vatten, L.J.; Laugsand, L.E.; Janszky, I. Insomnia and the Risk of Breast Cancer: The HUNT Study. Psychosom. Med. 2017, 79, 461–468. [Google Scholar] [CrossRef] [PubMed]
  81. Sigurdardottir, L.; Valdimarsdottir, U.A.; Mucci, L.; Fall, K.; Rider, J.R.; Schernerhammer, E.S.; Czeisler, C.A.; Launer, L.; Harris, T.B.; Stampfer, M.J. Insomnia among elderly men and risk of prostate cancer. J. Clin. Oncol. Off. J. Am. Soc. Clin. 2012, 30, 78. [Google Scholar] [CrossRef]
  82. Gapstur, S.M.; Diver, W.R.; Stevens, V.L.; Carter, B.D.; Teras, L.R.; Jacobs, E.J. Work schedule, sleep duration, insomnia, and risk of fatal prostate cancer. Am. J. Prev. Med. 2014, 46, S26–S33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Luojus, M.K.; Lehto, S.M.; Tolmunen, T.; Erkkilä, A.T.; Kauhanen, J. Sleep duration and incidence of lung cancer in age-ing men. BMC Public Health 2014, 14, 295–301. [Google Scholar] [CrossRef] [Green Version]
  84. Sturgeon, S.R.; Luisi, N.; Balasubramanian, R.; Reeves, K.W. Sleep duration and endometrial cancer risk. Cancer Causes Control. 2012, 23, 547–553. [Google Scholar] [CrossRef]
  85. Vogtmann, E.; Levitan, E.B.; Hale, L.; Shikany, J.M.; Shah, N.A.; Endeshaw, Y.; Lewis, C.E.; Manson, J.E.; Chlebowski, R.T. Association between sleep and breast cancer incidence among postmenopausal women in the Women’s Health Initiative. Sleep 2013, 36, 1437–1444. [Google Scholar] [CrossRef] [Green Version]
  86. Berisha, A.; Shutkind, K.; Borniger, J.C. Sleep Disruption and Cancer: Chicken or the Egg? Front. Neurosci. 2022, 16, 856235. [Google Scholar] [CrossRef]
  87. Hakim, F.; Wang, Y.; Zhang, S.X.; Zheng, J.; Yolcu, E.S.; Carreras, A.; Khalyfa, A.; Shirwan, H.; Almendros, I.; Gozal, D. Fragmented Sleep Accelerates Tumor Growth and Progression through Recruitment of Tumor-Associated Macrophages and TLR4 Signaling. Cancer Res. 2014, 74, 1329–1337. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Möller-Levet, C.S.; Archer, S.N.; Bucca, G.; Laing, E.E.; Slak, A.; Kabiljo, R.; Lo, J.C.Y.; Santhi, N.; von Schantz, M.; Smith, C.P.; et al. Effects of insufficient sleep on circadian rhythmicity and expression amplitude of the human blood transcriptome. Proc. Natl. Acad. Sci. USA 2013, 110, E1132–E1141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Haus, E.L.; Smolensky, M.H. Shift work and cancer risk: Potential mechanistic roles of circadian disruption, light at night, and sleep deprivation. Sleep Med. Rev. 2013, 17, 273–284. [Google Scholar] [CrossRef] [PubMed]
  90. Mazzoccoli, G.; Tarquini, R.; Durfort, T.; Francois, J.C. Chronodisruption in lung cancer and possible therapeutic approaches. Biomed. Pharmacother. 2011, 65, 500–508. [Google Scholar] [CrossRef]
  91. Fu, L.; Lee, C.C. The circadian clock: Pacemaker and tumour suppressor. Nat. Rev. Cancer 2003, 3, 350–361. [Google Scholar] [CrossRef] [PubMed]
  92. Cos, S.; González, A.; Martinez-Campa, C.; Mediavilla, M.D.; Alonso-González, C.; Sánchez-Barceló, E.J. Estrogen-signaling pathway: A link between breast cancer and melatonin oncostatic actions. Cancer Detect. Prev. 2006, 30, 118–128. [Google Scholar] [CrossRef] [PubMed]
  93. Bovbjerg, D.H. Circadian disruption and cancer: Sleep and immune regulation. Brain Behav. Immun. 2003, 17, S48–S50. [Google Scholar] [CrossRef]
  94. Guarino, V.; Castellone, M.D.; Avilla, E.; Melillo, R.M. Thyroid cancer and inflammation. Mol. Cell. Endocrinol. 2010, 321, 94–102. [Google Scholar] [CrossRef] [Green Version]
  95. Buman, M.P.; Phillips, B.A.; Youngstedt, S.D.; Kline, C.E.; Hirshkowitz, M. Does nighttime exercise really disturb sleep? Results from the 2013 National Sleep Foundation Sleep in America Poll. Sleep Med. 2014, 15, 755–761. [Google Scholar] [CrossRef]
  96. Phelps, K. The Cancer Recovery Guide; MacMillan Publishers: Sydney, NSW, Australia, 2015. [Google Scholar]
  97. Anand, P.; Kunnumakara, A.B.; Sundarum, C.; Harikumar, K.B.; Tharakan, S.T.; Lai, O.S.; Sung, B.; Aggarwal, B.B. Cancer is a preventable disease that requires major lifestyle changes. Pharm. Res. 2008, 25, 2097–2116. [Google Scholar] [CrossRef]
  98. Campbell and Campbell. The China Study; Wakefield Press: Adelaide, SA, Australia, 2007. [Google Scholar]
  99. Calle, E.E.; Kaaks, R. Overweight, obesity and cancer: Epidemiological evidence and proposed mechanisms. Nat. Rev. Cancer 2004, 4, 579–591. [Google Scholar] [CrossRef] [PubMed]
  100. National Cancer Institute Obesity and Cancer Risk Fact Sheet. Updated 5 April 2022. Available online: https://www.cancer.gov/about-cancer/causes-prevention/risk/obesity/obesity-fact-sheet (accessed on 7 January 2022).
  101. Sung, H.; Siegel, R.L.; Torre, L.A.; Pearson-Stuttard, J.; Islami, F.; Fedewa, S.A.; Sauer, A.G.; Shuval, K.; Gapstur, S.M.; Jacobs, E.J.; et al. Global patterns in excess body weight and the associated cancer burden. CA Cancer J. Clin. 2018, 69, 88–112. [Google Scholar] [CrossRef] [PubMed]
  102. Benetou, V.; Trichopoulou, A.; Orfanos, P.; Naska, A.; Lagiou, P.; Boffetta, P.; Trichopoulos, D. Conformity to traditional Mediterranean diet and cancer incidence: The Greek EPIC cohort. Br. J. Cancer 2008, 99, 191–195. [Google Scholar] [CrossRef] [PubMed]
  103. Trichopoulou, A.; Costacou, T.; Bamia, C.; Trichopoulos, D. Adherence to a Mediterranean Diet and Survival in a Greek Population. N. Engl. J. Med. 2003, 348, 2599–2608. [Google Scholar] [CrossRef] [Green Version]
  104. Casas, R.; Sacanella, E.; Estruch, R. The immune protective effect of the Mediterranean Diet against chronic low-grade inflammatory diseases. Endocr. Metab. Immune Disord. Drug Targets 2014, 14, 245–254. [Google Scholar] [CrossRef] [Green Version]
  105. Simopoulos, A.P. The importance of the ratio of omega-6/omega-3 essential fatty acids. Biomed. Pharm. 2002, 56, 365–379. [Google Scholar] [CrossRef]
  106. Simopoulos, A.P. Evolutionary aspects of diet: The omega-6/omega-3 ratio and the brain. Mol. Neurobiol. 2011, 44, 203. [Google Scholar] [CrossRef]
  107. Wilkinson, D. Can Food Be Medicine Against Cancer? Inspiring Publishers: Canberra, Australia, 2015.
  108. Jing, K.; Wu, T.; Lim, K. Omega-3 polyunsaturated fatty acids and cancer. Anticancer. Agents. Med. Chem. 2013, 13, 1162–1167. [Google Scholar] [CrossRef]
  109. Escrich, E.; Moral, R.; Solanas, M. Olive oil, an essential component of the Mediterrean diet, and breast cancer. Public Health Nutr. 2011, 14, 2323–2332. [Google Scholar] [CrossRef] [Green Version]
  110. Carroll, K.L.; Frugé, A.D.; Heslin, M.J.; Lipke, E.A.; Greene, M.W. Diet as a risk factor for early-onset colorectal adenoma and carcinoma: A systematic review. Front. Nutr. 2022, 9, 896330. [Google Scholar] [CrossRef]
  111. Cohen, J.H.; Kristal, A.; Stanford, J.L. Fruit and vegetable intakes and prostate cancer risk. Gynecol. Oncol. 2000, 92, 61–68. [Google Scholar] [CrossRef] [Green Version]
  112. Kwan, M.L.; Weltzien, E.; Kushi, L.H.; Castillo, A.; Slattery, M.L.; Caan, B.J. Dietary patterns and breast cancer recurrence and survival among women with early-stage breast cancer. J. Clin. Oncol. 2009, 27, 919–926. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Lin, P.-W.; Aronson, W.; Feedland, S.J. Nutrition, dietary interventions and prostate cancer: The latest evidence. BMC Med. 2015, 13, 3. [Google Scholar] [CrossRef]
  114. Rock, C.L.; Demark-Wahnefried, W. Nutrition and Survival After the Diagnosis of Breast Cancer: A Review of the Evidence. J. Clin. Oncol. 2002, 20, 3302–3316. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Rock, C.L.; Flatt, S.W.; Natarajan, L.; Thomson, C.A.; Bardwell, W.A.; Newman, V.A.; Hollenbach, K.A.; Jones, L.; Caan, B.J.; Pierce, J.P. Plasma Carotenoids and Recurrence-Free Survival in Women with a History of Breast Cancer. J. Clin. Oncol. 2005, 23, 6631–6638. [Google Scholar] [CrossRef] [PubMed]
  116. Steinmetz, K.A.; Kushi, L.H.; Bostick, R.M.; Folsum, A.R.; Potter, J.D. Vegetables, fruit, and colon cancer in the lowa Women’s Health Study. Am. J. Epidemiol. 1994, 139, 1–15. [Google Scholar] [CrossRef]
  117. Markellos, C.; Ourailidou, M.-E.; Gavriatopoulou, M.; Halvatsiotis, P.; Sergentanis, T.N.; Psaltopoulou, T. Olive oil intake and cancer risk: A systematic review and meta-analysis. PLoS ONE 2022, 17, e0261649. [Google Scholar] [CrossRef]
  118. Roberts, D.C.; Chidambaram, S.; Kinross, J.M. The role of the colonic microbiota and bile acids in colorectal cancer. Curr. Opin. Gastroenterol. 2022, 38, 179–188. [Google Scholar] [CrossRef]
  119. Hajishafiee, M.; Saneei, P.; Benisi-Kohansal, S.; Esmaillzadeh, A. Cereal fibre intake and risk of mortality from all causes, CVD, cancer and inflammatory diseases: A systematic review and meta-analysis of prospective cohort studies. Br. J. Nutr. 2016, 116, 343–352. [Google Scholar] [CrossRef]
  120. Reynolds, A.; Mann, J.; Cummings, J.; Winter, N.; Mete, E.; Te Morenga, L. Carbohydrate quality and human health: A series of systematic reviews and meta-analyses. Lancet 2019, 393, 1017. [Google Scholar] [CrossRef] [Green Version]
  121. Shanahan, F. Fiber man meets microbial man. Am. J. Clin. Nutr. 2014, 101, 1–2. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Zou, J.; Chassaing, B.; Singh, V.; Pellizzon, M.; Ricci, M.; Fythe, M.D.; Kumar, M.V.; Gewirtz, A.T. Fiber-Mediated Nourishment of Gut Microbiota Protects against Diet-Induced Obesity by Restoring IL-22-Mediated Colonic Health. Cell Host Microbe. 2017, 23, 41–53.e4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Bauer, S.R.; Hankinson, S.E.; Bertone-Johnstone, E.R.; Ding, E.L. Plasma vitamin D levels, menopause, and risk of breast cancer: Dose-response meta-analysis of prospective studies. Med. Baltim. 2013, 92, 123–131. [Google Scholar] [CrossRef] [PubMed]
  124. Gandini, S.; Boniol, M.; Haukka, J.; Byrnes, G.; Cox, B.; Sneyd, M.J.; Mullie, P.; Autier, P. Meta-analysis of observational studies of serum 25-hydroxyvitamin D levels and colorectal, breast and prostate cancer and colorectal adenoma. Int. J. Cancer 2011, 128, 1414–1424. [Google Scholar] [CrossRef]
  125. Gorham, E.D.; Garland, C.F.; Garland, F.C.; Grant, W.; Mohr, S.B.; Lipkin, M.; Newmark, H.L.; Giovannucci, E.; Wei, M.; Holick, M. Optimal Vitamin D Status for Colorectal Cancer Prevention: A Quantitative Meta Analysis. Am. J. Prev. Med. 2007, 32, 210–216. [Google Scholar] [CrossRef]
  126. Ma, Y.; Zhang, P.; Wang, F.; Yang, J.; Liu, Z.; Qin, H. Association Between Vitamin D and Risk of Colorectal Cancer: A Systematic Review of Prospective Studies. J. Clin. Oncol. 2011, 29, 3775–3782. [Google Scholar] [CrossRef]
  127. Bjelakovic, G.; Gluud, L.L.; Nikolova, D.; Whitfield, K.; Wetterslev, J.; Simonetti, R.G.; Bjelakovic, M.; Gluud, C. Vitamin D supplementation for prevention of mortality in adults. Cochrane Database Syst. Rev. 2014, CD007470. [Google Scholar] [CrossRef]
  128. Kim, Y.; Je, Y. Vitamin D intake, blood 25(OH)D levels, and breast cancer risk or mortality: A meta-analysis. Br. J. Cancer 2014, 110, 2772–2784. [Google Scholar] [CrossRef] [Green Version]
  129. Mohr, S.B.; Gorham, E.D.; Kim, J.; Hofflich, H.; Cuomo, R.E.; Garland, C.F. Could vitamin D sufficiency improve the survival of colorectal cancer patients? J. Steroid. Biochem. 2015, 148, 239–244. [Google Scholar] [CrossRef]
  130. Mohr, S.B.; Gorham, E.D.; Kim, J.; Hofflich, H.; Garland, C.F. Meta-analysis of vitamin D sufficiency for improving survival of patients with breast cancer. Anticancer Res. 2014, 34, 1163–1166. [Google Scholar]
  131. Zgaga, L.; Theodoratou, E.; Farrington, S.M.; Din, F.V.; Ooi, L.Y.; Glodzik, D.; Johnston, S.; Tenesa, A.; Campbell, H.; Dunlop, M.G. Plasma Vitamin D Concentration Influences Survival Outcome After a Diagnosis of Colorectal Cancer. J. Clin. Oncol. 2014, 32, 2430–2439. [Google Scholar] [CrossRef] [PubMed]
  132. Moukayed, M.; Grant, W.B. Molecular link between vitamin D and cancer prevention. Nutrients 2013, 5, 3993–4021. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Bikle, D.D. Vitamin D Metabolism, mechanisms of action, and clinical applications. Chem. Biol. 2014, 21, 319–329. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Díaz, G.D.; Paraskeva, C.; Thomas, M.G.; Binderup, L.; Hague, A. Apoptosis is induced by the active metabolite of vitamin D3 and its analogue EB1089 in colorectal adenoma and carcinoma cells: Possible implications for prevention and therapy. Cancer Res. 2000, 60, 2304–2312. [Google Scholar]
  135. Scaglione-Sewell, B.A.; Bissonnette, M.; Skarosi, S.; Abraham, C.; Brasitus, T.A. A vitamin D3 analog induces a G1-phase arrest in CaCo-2 cells by inhibiting cdk2 and cdk6: Roles of cyclin E, p21Waf1, and p27Kip1. Endocrinology 2000, 141, 3931–3939. [Google Scholar] [CrossRef]
  136. Chandler, P.D.; Chen, W.Y.; Ajala, O.N.; Hazra, A.; Cook, N.; Bubes, V.; Lee, I.-M.; Giovannucci, G.L.; Willett, W.; Buring, J.E. Effect of Vitamin D3 Supplements on development of advanced cancer: A secondary analysis of the VITAL randomized clinical trial. JAMA Netw Open 2020, 3, e2025850. [Google Scholar] [CrossRef]
  137. Urashima, M.; Ohdaira, H.; Akutsu, T.; Odaira, H.; Suzuki, Y.; Urashima, M. Effect of vitamin D supplementation on relapse-free survival among patients with digestive tract cancers: The AMATERASU randomized clinical trial. JAMA 2019, 321, 1361–1369. [Google Scholar] [CrossRef]
  138. Ng, K.; Nimeiri, H.S.; McCleary, N.J.; Abrams, T.A.; Yurgelun, M.B.; Cleary, J.M.; Rubinson, D.A.; Schrag, D.; Miksad, R.; Bullock, A.J.; et al. Effect of high-dose vs. standard-dose vitamin D3 supplementation on progression-free survival among patients with advanced or metastatic colorectal cancer: The SUNSHINE randomized clinical trial. JAMA 2019, 321, 1370–1379. [Google Scholar] [CrossRef] [Green Version]
  139. Keum, N.; Giovannucci, E. Vitamin D supplements and cancer incidence and mortality: A meta-analysis. Br. J. Cancer 2014, 111, 976–980. [Google Scholar] [CrossRef] [Green Version]
  140. Manson, J.E.; Cook, N.R.; Lee, I.M.; Cristen, W.; Bassuk, S.S.; Mora, S.; Gibson, H.; Gordon, D.; Copeland, T.; D’Agostino, D.; et al. Vitamin D supplements and prevention of cancer and cardiovascular disease. N. Engl. J. Med. 2019, 380, 33–44. [Google Scholar] [CrossRef]
  141. Shen, D.; Mao, W.; Liu, T. Sedentary behavior and incident cancer: A meta-analysis of prospective studies. PLoS ONE 2014, 9, e105709. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Lynch, B. Sedentary behaviour and cancer: A systematic review of the literature and proposed biological mechanisms. Cancer Epidemiol. Biomark. Prev. 2010, 19, 2691–2709. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Cong, Y.J.; Gan, Y.; Sun, H.L.; Deng, J.; Cao, S.Y.; Xu, X.; Lu, Z.X. Association of sedentary behaviour with colon and rectal cancer: A meta-analysis of observational studies. Br. J. Cancer 2014, 110, 817–826. [Google Scholar] [CrossRef] [Green Version]
  144. Meyerhardt, J.A.; Giovannucci, E.L.; Holmes, M.D.; Chan, A.T.; Chan, J.A.; Colditz, G.A.; Fuchs, G.S. Physical activity and survival after colorectal cancer diagnosis. J. Clin. Oncol. 2006, 24, 3527–3534. [Google Scholar] [CrossRef] [PubMed]
  145. Schmidt, M.E.; Chang-Claude, J.; Vrieling, A.; Seibold, P.; Heinz, J.; Obi, N.; Flesch-Janys, D.; Steindorf, K. Association of pre-diagnosis physical activity with recurrence and mortality among women with breast cancer. Int. J. Cancer 2013, 133, 1431–1441. [Google Scholar] [CrossRef]
  146. Hamer, M.; Stamatakis, E.; Saxton, J.M. The impact of physical activity on all-cause mortality in men and women after a cancer diagnosis. Cancer Causes Control. 2008, 20, 225–231. [Google Scholar] [CrossRef] [PubMed]
  147. Friedenreich, C.M.; Ryder-Burbidge, C.; McNeil, J. Physical activity, obesity and sedentary behavior in cancer etiology: Epidemiologic evidence and biologic mechanisms. Mol. Oncol. 2020, 15, 790–800. [Google Scholar] [CrossRef]
  148. Birks, S.; Peeters, A.; Backholer, K.; O’Brien, P.; Brown, W. A systematic review of the impact of weight loss on cancer incidence and mortality. Obes. Rev. 2012, 13, 868–891. [Google Scholar] [CrossRef]
  149. Jurdana, M. Physical activity and cancer risk. Actual knowledge and possible biological mechanisms. Radiol. Oncol. 2021, 55, 7–17. [Google Scholar] [CrossRef]
  150. Lahart, I.; Metsios, G.S.; Nevill, A.; Carmichael, A.R. Physical activity, risk of death and recurrence in breast cancer survivors: A systematic review and meta-analysis of epidemiological studies. Acta Oncol. 2015, 54, 635–654. [Google Scholar] [CrossRef]
  151. Moore, S.C.; Lee, I.-M.; Weiderpass, E.; Campbell, P.T.; Sampson, J.N.; Kitahara, C.M.; Keadle, S.K.; Arem, H.; de Gonzalez, A.B.; Hartge, P.; et al. Association of Leisure-Time Physical Activity With Risk of 26 Types of Cancer in 1.44 Million Adults. JAMA Intern. Med. 2016, 176, 816–825. [Google Scholar] [CrossRef] [PubMed]
  152. Swift DLMcGee, J.E.; Earnest, C.P.; Carlisle, E.; Nygard, M.; Johannsen, N.M. The effects of exercise and physical activity on weight loss and maintenance. Prog. Cardiovasc. Dis. 2018, 61, 206–213. [Google Scholar] [CrossRef] [PubMed]
  153. Metsios, G.S.; Moe, R.H.; Kitas, G.D. Exercise and inflammation. Best Pract. Res. Clin. Rheumatol. 2020, 34, 101504. [Google Scholar] [CrossRef]
  154. Zhong, D.; Li, Y.; Huang, Y.; Hong, X.; Li, J.; Jin, R. Molecular Mechanisms of Exercise on Cancer: A Bibliometrics Study and Visualization Analysis via CiteSpace. Front. Mol. Biosci. 2022, 8, 797902. [Google Scholar] [CrossRef] [PubMed]
  155. Dimeo, F.C. Effects of exercise on cancer-related fatigue. Cancer Relat. Fatigue New Dir. Res. 2001, 92, 1689–1693. [Google Scholar] [CrossRef]
  156. Campbell, K.L.; Winters-Stone, K.M.; Wiskemann, J.; May, A.M.; Schwartz, A.L.; Courneya, K.S.; Zucker, D.; Matthews, C.; Ligibel, J.; Gerber, L.; et al. Exercise guidelines for cancer survivors: Consensus statement from International Multidisciplinary Roundtable. Med. Sci. Sports Exerc. 2019, 51, 2375–2390. [Google Scholar] [CrossRef] [Green Version]
  157. Holmes, M.D.; Chen, W.Y.; Feskanich, D.; Kroenke, C.H.; Colditz, G.A. Physical Activity and Survival After Breast Cancer Diagnosis. Obstet. Gynecol. Surv. 2005, 60, 798–800. [Google Scholar] [CrossRef] [Green Version]
  158. Lee, I.-M.; Wolin, K.Y.; Freeman, S.E.; Sattlemair, J.; Sesso, H.D.; Sattelmair, J. Physical activity and survival after cancer diagnosis in men. J. Phys. Act. Health 2014, 11, 85–90. [Google Scholar] [CrossRef] [Green Version]
  159. Kim, J.-S.; Wilson, R.L.; Taaffe, D.R.; Galvão, D.A.; Gray, E.; Newton, R.U. Myokine expression and tumor-suppressive effect of serum after 12 wk of exercise in prostate cancer patients on ADT. Med. Sci. Sports Exerc. 2022, 54, 197–205. [Google Scholar] [CrossRef]
  160. Kim, J.-S.; Galvao, D.A.; Newton, R.U.; Gray, E.; Taaffe, D.R. Exercise-induced myokines and their effect on prostate cancer. Nat. Rev. Urol. 2021, 18, 519–542. [Google Scholar] [CrossRef]
  161. Hayes, S.C.; Spence, R.A.; Galvao, D.; Newton, R.U. Australian Association for Exercise and Sport Science position stand: Optimising cancer outcomes through exercise. J. Sci. Med. Sport 2009, 12, 428–434. [Google Scholar] [CrossRef] [Green Version]
  162. Wang, Q.; Zhou, W. Roles and molecular mechanisms of physical exercise in cancer prevention and treatment. J. Sport Health Sci. 2021, 10, 201–210. [Google Scholar] [CrossRef] [PubMed]
  163. Maggini, S.; Wintergerst, E.S.; Beveridge, S.; Hornig, D.H. Selected vitamins and trace elements support immune function by strengthening epithelial barriers and cellular and humoral immune responses. Br. J. Nutr. 2007, 98, S29–S35. [Google Scholar] [CrossRef]
  164. Chen, Y.-C.; Prabhu, K.S.; Mastro, A.M. Review: Is selenium a potential treatment for cancer metastasis? Nutrients 2013, 5, 1149–1168. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Perlmutter, D. Brain Maker; Yellow Kite Books: London, UK, 2015. [Google Scholar]
  166. Schwabe, R.F.; Jobin, C. The microbiome and cancer. Nat. Rev. Cancer 2013, 13, 800–812. [Google Scholar] [CrossRef] [PubMed]
  167. Al-Qadami, G.; Van Sebille, Y.; Bowen, J.; Wardill, H. Oral-gut microbiome axis in the pathogenesis of cancer treatment-induced oral mucositis. Front. Oral Health 2022, 3, 881949. [Google Scholar] [CrossRef] [PubMed]
  168. Deleemans, J.M.; Chleilat, F.; Reimer, R.A.; Henning, J.-W.; Baydoun, M.; Piedalue, K.-A.; McLennan, A.; Carlson, L.E. The chemo-gut study: Investigating the long-term effects of chemotherapy on gut microbiota, metabolic, immune, psychological and cognitive parameters in young adult Cancer survivors; study protocol. BMC Cancer 2019, 19, 1243. [Google Scholar] [CrossRef] [Green Version]
  169. Starr, P. Oral Nicotinamide Prevents Common Skin Cancers in High-Risk Patients, Reduces Costs. Am. Health Drug Benefits 2015, 8, 13–14. [Google Scholar]
  170. Dettman, I.; Meakin, C.; Allen, R. Co-infusing Glutathione and Vitamin C during cancer treatment: A reply. ACNEM J. 2012, 31, 8–11. [Google Scholar]
  171. Mikirova, N.; Jackson, J.; Riordan, N. The effect of high dose IV Vitamin C on plasma antioxidant capacity and level of oxidative stress in cancer patients and healthy subjects. Orthomol. Med. 2007, 22, 153–160. [Google Scholar]
  172. Shiota, M.; Yokomizo, A.; Naito, S. Oxidative stress and androgen receptor signaling in the development and progression of castration-resistant prostate cancer. Free. Radic. Biol. Med. 2011, 51, 1320–1328. [Google Scholar] [CrossRef] [PubMed]
  173. Shiota, M.; Song, Y.; Takeuchi, A.; Yokomizo, A.; Kashiwagi, E.; Kuroiwa, K.; Tatsugami, K.; Uchiumi, T.; Oda, Y.; Naito, S. Antioxidant therapy alleviates oxidative stress by androgen deprivation and prevents conversion from androgen dependent to castration resistant prostate cancer. J. Urol. 2012, 187, 707–714. [Google Scholar] [CrossRef] [PubMed]
  174. Valko, M.; Rhodes, C.J.; Moncol, J.; Izakovic, M.; Mazur, M. Free radicals, metals and antioxidants in oxidative stress-induced cancer. Chem. Biol. Interact. 2006, 160, 1–40. [Google Scholar] [CrossRef] [PubMed]
  175. Wybieralska, E.; Koza, M.; Sroka, J.; Czyż, J.; Madeja, Z. Ascorbic acid inhibits the migration of walker 256 carcinosarcoma cells. Cell. Mol. Biol. Lett. 2008, 13, 103–111. [Google Scholar] [CrossRef] [PubMed]
  176. Gonzalez, M.J.; Miranda-Massari, J.R.; Mora, E.M.; Guzman, A.; Riordan, N.H.; Casciari, J.J.; Jackson, J.A.; Roman-Franco, A. Orthomolecular oncology review: Ascorbic acid and cancer 25 years later. Integr. Cancer Ther. 2005, 4, 32–44. [Google Scholar] [CrossRef]
  177. Block, K.I.; Koch, A.C.; Mead, M.N.; Tothy, P.K.; Newman, R.A.; Gyllenhaal, C. Impact of antioxidant supplementation on chemotherapeutic efficacy: A systematic review of the evidence from randomized controlled trials. Cancer Treat. Rev. 2007, 33, 407–418. [Google Scholar] [CrossRef]
  178. Block, K.I.; Koch, A.C.; Mead, M.N.; Tothy, P.K.; Newman, R.A.; Gyllenhaal, C. Impact of antioxidant supplementation on chemotherapeutic toxicity: A systematic review of the evidence from randomized controlled trials. Int. J. Cancer 2008, 123, 1227–1239. [Google Scholar] [CrossRef]
  179. Bohm, S.; Oriana, S.; Spatti, G.; Di Re, F.; Breasciani, g.; Pirovano, C.; Grosso, I.; Martini, C.; Caraceni, A.; Pilotti, S.; et al. Dose intensification of platinum compounds with glutathione protection as induction chemotherapy for advanced ovarian cancer. Oncology 1999, 57, 115–120. [Google Scholar] [CrossRef]
  180. Cascinu, S.; Catalano, V.; Cordella, L.; Labianca, R.; Giordani, P.; Baldelli, A.M.; Beretta, G.D.; Ubiali, E.; Catalano, G. Neuroprotective effect of reduced glutathione on oxaliplatin-based chemotherapy in advanced colorectal cancer: A randomized, double-blind, placebo-controlled trial. J. Clin. Oncol. 2002, 20, 3478–3483. [Google Scholar] [CrossRef]
  181. Böttger, F.; Vallés-Martí, A.; Cahn, L.; Jimenez, C.R. High-dose intravenous vitamin C, a promising multi-targeting agent in the treatment of cancer. J. Exp. Clin. Cancer Res. 2021, 40, 343. [Google Scholar] [CrossRef]
  182. González, M.J.; Rosario-Pérez, G.; Guzmán, A.M.; Miranda-Massari, J.R.; Duconge, J.; Lavergne, J.; Fernandez, N.; Ortiz, N.; Quintero, A.; Mikirova, N.; et al. Mitochondria, Energy and Cancer: The Relationship with Ascorbic Acid. J. Orthomol. Med. Off. J. Acad. Orthomol. Med. 2010, 25, 29–38. [Google Scholar]
  183. Vollbracht, C.; Schneider, B.; Leendert, V.; Weiss, G.; Auerbach, L.; Beuth, J. Intravenous vitamin C administration improves quality of life in breast cancer patients during chemo-/radiation therapy and aftercare: Results of a retrospective, multicentre, epidemiological cohort study in Germany. In Vivo 2011, 25, 983–990. [Google Scholar]
  184. Travica, N.; Ried, K.; Hudson, I.; Scholey, A.; Pipingas, A.; Sali, A. The effects of surgery on plasma/serum vitamin C concentrations: A systematic review and meta-analysis. Br. J. Nutr. 2020, 127, 1–15. [Google Scholar] [CrossRef] [PubMed]
  185. Ravindran, J.; Prasad, S.; Aggarwal, B.B. Curcumin and Cancer Cells: How Many Ways Can Curry Kill Tumor Cells Selectively? AAPS J. 2009, 11, 495–510. [Google Scholar] [CrossRef] [PubMed]
  186. Wilken, R.; Veena, M.S.; Wang, M.B.; Srivatsan, E.S. Curcumin: A review of anti-cancer properties and therapeutic activity in head and neck squamous cell carcinoma. Mol. Cancer 2011, 10, 12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Li, Z.; Zhang, L.; Wang, H.; Ma, J.-X.; Sun, J.-Z. Curcumin inhibits lung cancer progression and metastasis through induction of FOXO1. Tumor. Biol. 2014, 35, 111. [Google Scholar] [CrossRef]
  188. Zhou, D.Y.; Ding, N.; Du, Z.Y.; Cui, X.X.; Wang, H.; Wei, X.C.; Conney, A.H.; Zhang, K.; Zheng, X. Curcumin analogues with high activity for inhibiting human prostate cancer cell growth and androgen receptor activation. Mol. Med. Rep. 2014, 10, 1315–1322. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  189. Dorai, T.; Diouri, J.; O’Shea, O.; Doty, S.B. Curcumin inhibits prostate cancer bone metastasis by up-regulating bone morphogenic protein-7 in vivo. J. Cancer Ther. 2014, 5, 369–386. [Google Scholar] [CrossRef] [Green Version]
  190. Du, Y.; Long, Q.; Zhang, L.; Shi, Y.; Liu, X.; Li, X.; Guan, B.; Tian, Y.; Wang, X.; Li, L.; et al. Curcumin inhibits cancer-associated fibroblast-driven prostate cancer invasion through MAOA/mTOR/HIF-1α signaling. Int. J. Oncol. 2015, 47, 2064–2072. [Google Scholar] [CrossRef] [Green Version]
  191. Aggarwal, B.B.; Shishodia, S.; Takada, Y.; Banerjee, S.; Newman, R.A.; Buesos-Ramos, C.E.; Price, J.E. Curcumin suppresses the paclitaxel-induced nuclear factor-kappaB pathway in breast cancer cells and inhibits lung metastasis of human breast cancer in nude mice. Clin. Cancer Res. 2005, 11, 7490–7498. [Google Scholar] [CrossRef]
  192. Huang, M.T.; Wang, Z.Y.; Georgiadis, C.A.; Laskin, J.D.; Conney, A.H. Inhibitory effects of curcumin on tumor initiation by benzo[a]pyrene and 7, 12 dimethylbenz[a]anthracene. Carcinogenesis 1992, 13, 2183–2186. [Google Scholar] [CrossRef] [PubMed]
  193. Lopresti, A.L.; Drummond, P.D. Efficacy of curcumin, and a saffron/curcumin combination for the treatment of major depression: A randomised, double-blind, placebo-controlled study. J. Affect. Disord. 2017, 207, 188–196. [Google Scholar] [CrossRef] [PubMed]
  194. Lopresti, A.L.; Maes, M.; Maker, G.L.; Hood, S.D.; Drummond, P.D. Curcumin for the treatment of major depression: A randomised, double-blind, placebo controlled study. J. Affect. Disord. 2014, 167, 368–375. [Google Scholar] [CrossRef] [Green Version]
  195. Hao, E.; Mukhopadhyay, P.; Cao, Z.; Erdélyi, K.; Holovac, E.; Liaudet, L.; Lee, W.-S.; Haskó, G.; Mechoulam, R.; Pacher, P. Cannabidiol Protects against Doxorubicin-Induced Cardiomyopathy by Modulating Mitochondrial Function and Biogenesis. Mol. Med. 2015, 21, 38–45. [Google Scholar] [CrossRef] [PubMed]
  196. Pan, H.; Mukhopadhyay, P.; Rajesh, M.; Patel, V.; Mukhopadhyay, B.; Gao, B.; Haskó, G.; Pacher, P. Cannabidiol Attenuates Cisplatin-Induced Nephrotoxicity by Decreasing Oxidative/Nitrosative Stress, Inflammation, and Cell Death. J. Pharmacol. Exp. Ther. 2008, 328, 708–714. [Google Scholar] [CrossRef] [Green Version]
  197. Ward, S.J.; McAllister, S.D.; Kawamura, R.; Murase, Y.; Neelakantan, H.; Walker, E.A. Cannabidiol inhibits paclitaxel-induced neuropathic pain through 5-HT 1A receptors without diminishing nervous system function or chemotherapy efficacy. J. Cereb. Blood Flow. Metab. 2014, 171, 636–645. [Google Scholar] [CrossRef] [Green Version]
  198. Taha, T.; Meiri, D.; Talhamy, S.; Wollner, M.; Peer, A.; Bar-Sela, G. Cannabis Impacts Tumor Response Rate to Nivolumab in Patients with Advanced Malignancies. Oncol. 2019, 24, 549–554. [Google Scholar] [CrossRef] [Green Version]
  199. Cao, Y.; Xu, X.; Liu, S.; Huang, L.; Gu, J. Ganoderma: A cancer immunotherapy review. Front. Pharmacol. 2018, 9, 1217. [Google Scholar] [CrossRef] [Green Version]
  200. Wachtel-Galor, S.; Yuen, J.; Buswell, J.A.; Benzie, I.F.F. Ganoderma lucidum (Lingzhi or Reishi): A medicinal mushroom. In Herbal Medicine: Biomolecular and Clinical Aspects, 2nd ed.; CRC Press: Boca Raton, FL, USA; Taylor & Francis: Boca Raton, FL, USA, 2011. [Google Scholar]
  201. Liu, J.; Shimizu, K.; Konishi, F.; Noda, K.; Kumamoto, S.; Kurashiki, K.; Kondo, R. Anti-androgenic activities of the triterpenoids fraction of Ganoderma lucidum. Food Chem. 2007, 100, 1691–1696. [Google Scholar] [CrossRef]
  202. Liu, J.; Shimizu, K.; Konishi, F.; Kumamoto, S.; Kondo, R. The anti-androgen effect of ganoderol B isolated from the fruiting body of Ganoderma lucidum. Bioorganic Med. Chem. 2007, 15, 4966–4972. [Google Scholar] [CrossRef]
  203. Gao, Y.; Zhou, S.; Jiang, W.; Huang, M.; Dai, X. Effects of ganopoly (a Ganoderma lucidum polysaccharide extract) on the immune functions in advanced-stage cancer patients. Immunol. Invest. 2003, 32, 201–215. [Google Scholar] [CrossRef] [PubMed]
  204. Jin, X.; Beguerie, J.R.; Sze, D.M.-Y.; Chan, G.C. Ganoderma lucidum (Reishi mushroom) for cancer treatment. Cochrane Database Syst. Rev. 2012, CD007731. [Google Scholar] [CrossRef] [PubMed]
  205. Zhu, L.; Li, L.; Li, Y.; Wang, J.; Wang, Q. Chinese Herbal Medicine as an Adjunctive Therapy for Breast Cancer: A Systematic Review and Meta-Analysis. Evid. Based Complement. Altern. Med. 2016, 2016, 1–17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  206. Li, S.; So, T.-H.; Tang, G.; Tan, H.-Y.; Wang, N.; Ng, B.F.L.; Chan, C.K.W.; Yu, E.C.-L.; Feng, Y. Chinese herbal medicine for reducing chemotherapy-associated side-effects in breast cancer patients: A systematic review and meta-analysis. Front. Oncol. 2020, 10, 599073. [Google Scholar] [CrossRef]
  207. O’Brien, K.; Weber, D. Insomnia in Chinese medicine: The Heart of the Matter. JACM 2016, 22, 684–694. [Google Scholar] [CrossRef] [PubMed]
  208. Weber, D.; O’Brien, K. Cancer and Cancer-Related Fatigue and the Interrelationships With Depression, Stress, and Inflammation. J. Evid. Based Integr. Med. 2016, 22, 502–512. [Google Scholar] [CrossRef]
  209. Lu, W.; Dean-Clower, E.; Doherty-Gilman, A.; Rosenthal, D.S. The value of acupuncture in cancer care. Hematol. Oncol. Clin. North Am. 2008, 22, 631–648. [Google Scholar] [CrossRef] [Green Version]
  210. Meng, Z.; Barcia, M.K.; Hu, C.; Chiang, J.; Chambers, M.; Rosenthal, D.I.; Peng, H.; Zhang, Y.; Zhao, Q.; Zhao, G.; et al. Randomized controlled trial of acupuncture for prevention of radiation-induced xerostomia among patients with nasopharyngeal carcinoma. Cancer 2012, 118, 3337–3344. [Google Scholar] [CrossRef] [Green Version]
  211. Shen, J.; Wenger, N.; Glaspy, J.; Hays, R.D.; Albert, P.S.; Choi, C.; Shekelle, P.G. Electroacupuncture for control of myeloablative chemotherapy-induced emesis: A randomized controlled trial. JAMA 2000, 284, 2755–2761. [Google Scholar] [CrossRef] [Green Version]
  212. Tas, D.; Uncu, D.; Sendur, M.A.; Koca, N.; Zengin, N. Acupuncture as a complementary treatment for cancer patients receiving chemotherapy. Asian Pac. J. Cancer Prev. 2014, 15, 3139–3144. [Google Scholar] [CrossRef]
  213. Yang, J.; Wahner-Roedler, D.L.; Zhou, X.A.; Johnson, L.; Do, A.; Pachman, D.R.; Chon, T.Y.; Salinas, M.; Millstine, D.A.; Bauer, B. Acupuncture for palliative cancer pain management: Systematic review. BMJ Support. Palliat. Care 2021, 11, 264–270. [Google Scholar] [CrossRef]
  214. Zhang, Y.; Lin, L.; Li, H.; Hu, Y.; Tian, L. Effects of acupuncture on cancer-related fatigue: A meta-analysis. Support. Care Cancer 2018, 26, 415–425. [Google Scholar] [CrossRef] [PubMed]
  215. Cancer Council NSW. Massage. Available online: https://www.cancercouncil.com.au/cancer-information/living-well/complementary-therapies/body-based-practices/massage/ (accessed on 12 October 2022).
  216. Campeau, M.P.; Gaboriault, R.; Drapeau, M.; Nguyen, T.V.; Roy, I.; Fortin, B.; Marois, M.; Nguyen-Tan, P.F. The impact of massage therapy on anxiety levels in patients undergoing radiation therapy: A randomized controlled trial. J Soc Integr. Oncol. 2007, 5, 133–138. [Google Scholar] [CrossRef] [PubMed]
  217. Khiewkhern, S.; Promthet, S.; Sukprasert, A.; Eunhpinitpong, W.; Bradshaw, P. Effectiveness of Aromatherapy with Light Thai Massage for Cellular Immunity Improvement in Colorectal Cancer Patients Receiving Chemotherapy. Asian Pac. J. Cancer Prev. 2013, 14, 3903–3907. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  218. Sagar, S.; Dryden, T.; Wong, R.K. Massage Therapy for Cancer Patients: A Reciprocal Relationship between Body and Mind. Curr. Oncol. 2007, 14, 45–56. [Google Scholar] [CrossRef] [Green Version]
  219. McIntyre, J.; Conroy, S.; Avery, A.; Corns, H.; Choonara, I. Unlicensed and off label prescribing of drugs in general practice. Arch. Dis. Child. 2000, 83, 498–501. [Google Scholar] [CrossRef]
  220. Gupta, S.K.; Nayak, R.P. Off-label use of medicine: Perspective of physicians, patients, pharmaceutical companies and regulatory authorities. J. Pharmacol. Pharmacother. 2014, 5, 88–92. [Google Scholar] [CrossRef] [Green Version]
  221. Soeters, P.B.; Grimble, R.F. The conditional role of inflammation in pregnancy and cancer. Clin. Nutr. 2013, 32, 460–465. [Google Scholar] [CrossRef]
  222. Heckman-Stoddard, B.M.; DeCensi, A.; Sahasrabuddhe, V.V.; Ford, L.G. Repurposing metformin for the prevention of cancer and cancer recurrence. Diabetologia 2017, 60, 1639–1647. [Google Scholar] [CrossRef] [Green Version]
  223. Chae, Y.K.; Arya, A.; Malecek, M.-K.; Shin, D.S.; Carneiro, B.; Chandra, S.; Kaplan, J.; Kalyan, A.; Altman, J.K.; Platanias, L.; et al. Repurposing metformin for cancer treatment: Current clinical studies. Oncotarget 2016, 7, 40767–40780. [Google Scholar] [CrossRef]
  224. Zhang, Z.-J.; Li, S. The prognostic value of metformin for cancer patients with concurrent diabetes: A systematic review and meta-analysis. Diabetes Obes. Metab. 2014, 16, 707–710. [Google Scholar] [CrossRef] [PubMed]
  225. Liubchenko, K.; Kordbacheh, K.; Khajehdehi, N.; Visnjevac, T. Naltrexone’s impact on cancer progression and mortality: A systematic review of studies in humans, animal models, and cell cultures. Adv. Ther. 2021, 38, 904–924. [Google Scholar] [CrossRef] [PubMed]
  226. Farhood, B.; Goradel, N.H.; Mortezaee, K.; Khanlarkhani, N.; Salehi, E.; Nashtaei, M.S.; Mirtavoos-Mahyari, H.; Motevaseli, E.; Shabeeb, D.; Musa, A.E.; et al. Melatonin as an adjuvant in radiotherapy for radioprotection and radiosensitization. Clin. Transl. Oncol. 2018, 21, 268–279. [Google Scholar] [CrossRef] [PubMed]
  227. Latocha, M.; Zyrek, L. Anti-cancer properties of dichloroacetate. Pol. Merkur. Lekarski. 2022, 50, 145–147. [Google Scholar] [PubMed]
  228. Wise, D.R.; Thompson, C.B. Glutamine addiction: A new therapeutic target in cancer. Trends Biochem. Sci. 2010, 35, 427–433. [Google Scholar] [CrossRef] [Green Version]
  229. Iannitti, T.; Palmieri, B. Clinical and experimental applications of sodium phenylbutyrate. Drugs R D 2011, 11, 227–249. [Google Scholar] [CrossRef]
  230. Gore, S.D.A.; Carducci, M. Modifying histones to tame cancer: Clinical development of sodium phenylbutyrate and other histone deacetylase inhibitors. Expert Opin. Investig. Drugs 2000, 9, 2923–2934. [Google Scholar] [CrossRef]
  231. Meade, B.; Sali, A.; Stephens, A.; Rainczuk, A.; Murphy, D.L. An investigation of metronomic photodynamic therapies for local prostate cancer: Supplementary fluorescent photography and proteomic studies, as measurable parameters of this treatment. J. Cancer Sci. Therap. 2018, 1, 8. [Google Scholar]
  232. Dery, M. Pharmacists Key to Safe Complementary Medicine Use, Australian Pharmacist. 2019. Available online: https://www.australianpharmacist.com.au/pharmacists-key-to-ensuring-safety-with-complementary-medicine/ (accessed on 22 March 2019).
  233. Gurley, B.; Fifer, E.; Gardner, Z. Pharmacokinetic Herb-Drug Interactions (Part 2): Drug Interactions Involving Popular Botanical Dietary Supplements and Their Clinical Relevance. Planta Med. 2012, 78, 1490–1514. [Google Scholar] [CrossRef]
  234. Izzo, A.A.; Ernst, E. Interactions between herbal medicines and prescribed drugs. Drugs 2009, 69, 1777–1798. [Google Scholar] [CrossRef]
  235. Yue, Q.Y.; Strandell, J.; Myrberg, O. Concomitant use of glucosamine may potentiate the effect of warfarin. Drug Saf. 2006, 29, 911–1010. [Google Scholar] [CrossRef]
  236. Tsai, Y.-T.; Lai, J.-N.; Wu, C.-T. The use of Chinese herbal products and its use on tamoxifam-induced endometrial cancer risk among female breast cancer patients: A population based study. J. Ethnopharm. 2014, 155, 1256–1262. [Google Scholar] [CrossRef]
  237. Jou, J.; Johnson, P.J. Nondisclosure of complementary and alternative medicine use to primary care physicians findings from the 2012 National Health Interview Survey. JAMA Intern. Med. 2016, 176, 545–546. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Pathomechanisms by which stress can contribute to cancer (figure reprinted with permission from [12]).
Figure 1. Pathomechanisms by which stress can contribute to cancer (figure reprinted with permission from [12]).
Cancers 14 05933 g001
Table 1. Examples of stress reduction therapies useful for cancer patients.
Table 1. Examples of stress reduction therapies useful for cancer patients.
Type of TherapySome Key BenefitsReferences
MeditationSignificantly less tumours compared with non-meditators; fewer admissions for cancer; decreased mortality from cancer; cancer regression [54,55,56]
Tai Chi and Qi GongPositive effects on cancer-specific quality of life, as well as fatigue, anxiety, immune function and cortisol levels[57]
Pet therapySignificant improvements in social and emotional wellbeing despite high symptom burden and expected decreases in physical and functional wellbeing associated with radiation-chemotherapy treatment [58]; pet therapy during chemotherapy associated with significant decrease in depression and increase in oxygen saturation [44][58,59]
Art therapyDecreased anxiety, depression and pain in patients with cancer; decreasing anxiety in those receiving cancer therapy; improvement in wellbeing[60,61,62,63,64]
Music therapyReduction of anxiety, pain, fatigue and improved mood and quality of life[65,66]
Writing therapyImproved quality of life[67]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

O’Brien, K.; Ried, K.; Binjemain, T.; Sali, A. Integrative Approaches to the Treatment of Cancer. Cancers 2022, 14, 5933. https://doi.org/10.3390/cancers14235933

AMA Style

O’Brien K, Ried K, Binjemain T, Sali A. Integrative Approaches to the Treatment of Cancer. Cancers. 2022; 14(23):5933. https://doi.org/10.3390/cancers14235933

Chicago/Turabian Style

O’Brien, Kylie, Karin Ried, Taufiq Binjemain, and Avni Sali. 2022. "Integrative Approaches to the Treatment of Cancer" Cancers 14, no. 23: 5933. https://doi.org/10.3390/cancers14235933

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop