Next Article in Journal
Long Non-Coding RNAs in Psoriasis and Cutaneous Squamous Cell Carcinoma
Previous Article in Journal
Factors Associated with Clinically Important Changes in Quality of Life of Heart Failure Patients: The QUALIFIER Prospective Cohort Study
Previous Article in Special Issue
Management of Bleeding, Thrombotic and Pregnancy-Related Complications in Women with Myeloproliferative Neoplasms: A Case-Based Review Focusing on Sex-Specific Challenges
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Management of Myeloproliferative Neoplasms: An Integrative Approach

by
Francesca Andreazzoli
1,2,*,
Ilana Levy Yurkovski
3,4,
Krisstina Gowin
5,† and
Massimo Bonucci
2,†
1
Hematology Department, North West Tuscany Hospital, 55041 Lucca, Italy
2
ARTOI Foundation, Via Ludovico Micara, 73, 00165 Rome, Italy
3
Hematology Unit, Bnai Zion Medical Center, Haifa 3339419, Israel
4
Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa 3200003, Israel
5
Departments of Supportive Care and Hematology Hematopoietic Transplant, City of Hope Comprehensive Cancer Center, Irvine, CA 91010, USA
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
J. Clin. Med. 2025, 14(14), 5080; https://doi.org/10.3390/jcm14145080
Submission received: 10 May 2025 / Revised: 13 July 2025 / Accepted: 15 July 2025 / Published: 17 July 2025

Abstract

Myeloproliferative neoplasms (MPNs) are chronic blood cancers characterized by overproduction of blood cells, leading to increased thrombotic and ischemic risk. Patients frequently experience symptoms including fatigue, abdominal discomfort, and complications from thrombotic events, which significantly impact the quality of life (QoL). Many patients inquire about complementary and integrative medicine (CIM) approaches, including nutritional interventions and supplements, creating opportunities for healthcare providers to engage in meaningful discussions guided by the principle of safety. This review examines the current evidence for integrative approaches in MPN management, focusing on nutrition, microbiota, supplements, mind–body techniques, and acupuncture. We analyze the available data on anti-inflammatory interventions, QoL improvement strategies, and treatment tolerance enhancement. The review provides clinicians with evidence-based guidance for safely integrating complementary therapeutic approaches with conventional MPN treatment. This integrative approach represents an opportunity to develop more comprehensive and personalized therapeutic paradigms in hematology while ensuring that complementary interventions serve as adjuncts to evidence-based medical treatment.

1. Introduction

Myeloproliferative neoplasms (MPNs), including polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF), are clonal hematopoietic stem cell disorders characterized by dysregulated hematopoiesis, chronic inflammation, and a variable risk of progression to acute leukemia [1,2]. Although therapeutic advancements have led to improved disease control and extended survival in some cases, MPNs remain incurable for most patients [3].
A central challenge in the management of MPNs is the high burden of physical and psychological symptoms, such as fatigue, pruritus, sleep disturbance, early satiety, mood disorders, and cognitive impairment. These symptoms are prevalent across disease subtypes and stages, often persist despite pharmacologic therapy, and significantly impair patients’ quality of life [4,5,6]. Conventional treatments, particularly the use of JAK inhibitors, while essential, frequently offer incomplete relief, and their side effects may further contribute to symptom distress [7].
In response, many patients turn to CIM to manage symptoms, enhance resilience—defined as the ability to adapt to and cope with the chronic and unpredictable nature of myeloproliferative neoplasms and their associated psychosocial stressors—and regain a sense of control. While resilience is frequently measured in cancer populations using general validated tools, such as the Connor–Davidson Resilience Scale [8], to our knowledge, no disease-specific definition or measurement of resilience currently exists for patients with MPNs. In the Survey of Integrative Medicine in Myeloproliferative Neoplasms (SIMM) study, over 50% of MPN patients reported using at least one CIM modality—most commonly aerobic activity, yoga, meditation, and acupuncture [9]. However, CIM use is often undisclosed to treating physicians, raising concerns about fragmented care, safety, and a lack of evidence-based integration into clinical practice [10].
Integrative oncology (IO) and integrative hematology (IH) are evolving frameworks that combine CIM with conventional care to address unmet supportive needs [11,12]. When delivered within a coordinated and scientifically informed framework, this approach has the potential to address the multifaceted symptom burden of MPNs and to support holistic patient care [13]. This evolving field, encompassing mind–body practices, lifestyle modification, nutritional strategies, and supportive interventions, warrants deeper exploration in the context of hematologic malignancies [14].
This narrative review aims to synthesize the current evidence surrounding integrative medicine in MPNs. Unfortunately, as often occurs in this research field, the data we have available are often preclinical, and very few clinical studies exist, with randomized trials being completely lacking. Although this may be frustrating, particularly considering the great demand from patients for an integrative approach, the collected data serve primarily as a starting point for building recommendations that will need to be expanded with further clinical studies. Another aspect to consider is that, especially in light of new targeted therapies and possible drug interactions, it is very important to understand which substances may have a rationale and which could instead be potentially harmful to the patient. Exploring these aspects, together with the link to nutrition, lifestyle, and mind–body techniques, is the purpose of this review (Figure 1).

2. Nutrition, Microbiota, and MPNs

While the relationship between food and nutrition is increasingly evident for solid tumors [15], evidence on the impact of nutrition in hematology is lacking.
Research is early but evolving in hematologic disorders; for example, an interesting link between multiple myeloma (MM) and nutrition is emerging. While data are preliminary, a Mediterranean-style diet with a drastic reduction in animal and processed products is suggested to reduce the evolution from monoclonal gammopathy of undetermined significance (MGUS) to MM [16].
In MPNs, the data are limited regarding the impact of nutrition on outcomes. Nutrition interventions are particularly interesting in this population due to the chronicity of the disease and the possibility for cardiovascular event risk reduction. Patients who adopt a healthy eating pattern may modulate cardiovascular risk and potentially even reduce inflammation and consequent fibrosis. Moreover, patients themselves often ask the hematologist what they should eat, wanting to take partial active control over their own health, and if the hematologist provides comprehensive answers, patients will be more adherent to these recommendations [17].

2.1. Inflammation

Inflammation, in fact, has been recognized in MPNs as a critical element of disease pathogenesis, with increased inflammatory cytokines being a hallmark of the disease and contributing to symptomatology and disease progression [18,19]; the cytokine profile differs in the different disease subtypes, demonstrating the role they play in guiding toward a specific phenotype. The main cytokines involved appear to be IL-1β, IL6, IL8, VEGF, and TGFβ. Among these, TGFβ in particular is involved in the induction of bone marrow fibrosis. The JAK2 (V617F) mutation itself, pathognomonic of MPNs, is associated with constitutive activation of the NLRP3 inflammasome, leading to a chronic inflammatory state. Finally, inflammasome activation induces the same hematopoietic stem and progenitor cells mutated in MPNs to produce greater amounts of IL-1β, contributing to a pro-inflammatory bone marrow microenvironment. This chronic inflammatory state favors clonal expansion of mutated cells, creating a vicious cycle that perpetuates the disease [20]. Finally, age and aging processes are considered pro-inflammatory states accompanied by high levels of pro-inflammatory cytokines; this could contribute to the pathogenesis of MPNs, whose incidence increases with age [21].
Targeting inflammation-related pathways is the effect of many targeted therapies, starting from JAK inhibitors to the more recent BET inhibitors [22]. Since many patients cannot benefit from pharmacological therapy because, for example, they do not meet the criteria to start it or due to intolerance, other approaches aimed at reducing the inflammatory burden could potentially reduce the symptom burden and also slow the course of the disease by extinguishing its trigger.
Unlike the common Western diet, whose inflammatory impact is documented, a Mediterranean diet has the ability to reduce inflammation [23] but also improve the cardiovascular profile of patients whose thrombotic risk is already intrinsic to the disease [24]. Moreover, a diet low in animal proteins and rich in vegetables, legumes, nuts, whole grains, and oily fish has a favorable impact on the hematological patient through mechanisms other than anti-inflammatory ones, such as the reduction in IGF1, the reduction in glucose intake, and the anti-tumor properties of phytochemicals contained in the food itself [25].

2.2. IGF1 and Insulin

The role of IGF1 in MPNS has, to date, been little studied, but a connection has nevertheless been established. This growth factor is involved both in normal physiological erythropoiesis as well as in erythropoiesis caused by excessive activation of the JAK2 protein; inhibition of IGF1 receptors prevents Epo-hypersensitive erythroid cell colony formation in preclinical models [26]. The cross-link between JAK2 and IGF1 is well known; IGF1 promotes cell growth by activating the PI3K/AKT and MAPK pathways that overlap with those of JAK/STAT, amplifying them. Moreover, IGF1 can directly activate JAK2, and JAK2 hyperexpression seems to be linked to the greater expression of IGF1 receptors by PV progenitors [27]. IGF1 levels could potentially be monitored in patients with myeloproliferative neoplasms as a biomarker for nutritional intervention efficacy, particularly given that recent meta-analytic evidence demonstrates an association between IGF1 level ranges and all-cause mortality, with a specific 120–160 ng/mL range being associated with the lowest mortality. Serum IGF1 concentrations are modifiable through dietary changes, particularly through reducing animal protein and sugar intake while adopting plant-based dietary patterns [28]. The identification of this optimal IGF1 range provides a concrete therapeutic target for interventions; however, the clinical utility and cost effectiveness of serial IGF1 measurements in this specific patient population require further investigation to determine whether achieving and maintaining IGF1 levels within the 120–160 ng/mL range translates into improved quality-of-life outcomes and survival benefits in myeloproliferative neoplasm patients. Additionally, the potential use of IGF1-lowering agents, such as metformin, warrants evaluation in future studies, contingent upon establishing the clinical relevance of IGF1 modulation within this target range for MPNs management [29,30]. Pathological cells in MPNs, characterized by JAK2 mutation, exhibit enhanced glucose uptake and increased glycolytic activity compared to normal hematopoietic cells [31,32].
Furthermore, the JAK2/STAT5 pathway, once constitutionally activated, increases the expression of the inducible rate-limiting enzyme 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3), allowing greater use of circulating glucose, useful for its metabolism and survival [33]. This could support the rationale for a low-glycemic-load diet in these patients.
The Nutrient Trial has demonstrated the feasibility and adherence to a Mediterranean diet by patients with MPNs, with 80% of patients being able to maintain good adherence to the proposed dietary pattern. The authors also showed that, in the group following a Mediterranean diet compared to the one following the standard US Dietary Guidelines for Americans (USDA), there was an improvement in symptoms after only 10 weeks of intervention. The lack of changes in inflammatory cytokine levels was attributed to the small sample size and short intervention period [34].

2.3. Obesity

Obesity is involved in the pathogenesis of MPNs through many mechanisms. First of all, some evidence shows that it represents a risk factor for MPNs, particularly in patients with ET [35,36]. Obesity also contributes to the production of inflammatory molecules such as interleukin-6 (IL6) and tumor necrosis factor-α (TNFα), favoring the creation of an environment useful for MPNs’ progression [37]. The evidence that MPNs, even in the absence of obesity, generate a specific inflammatory cytokine signature, combined with the known inflammatory effects of obesity, suggests a potential for synergistic inflammatory effects when both conditions coexist [38,39]. However, the relative contributions of hematologic disease versus adiposity to symptom burden, particularly fatigue, remain to be elucidated through studies with appropriate BMI-stratified analyses.
While fatigue is reported by almost 90% of obese patients with MPNs [39], the lack of comparative data from non-obese MPN cohorts limits our ability to determine whether this high prevalence is primarily attributable to the underlying neoplasm, obesity-related factors, or their interaction; the cytokine most responsible for fatigue would be TNFα, which, in fact, is increased both in patients with MPN and in the general obese population [40,41]. The altered production of leptin and adiponectin could compromise the bone marrow microenvironment and also normal hematopoiesis [42]. Obese patients also present an altered insulin and metabolic pattern characterized by insulin resistance and increased IGF1 [43]; the latter in particular could contribute to the triggering or progression of altered molecular signals, as explained above, responsible for disease progression [28].
Finally, the obese phenotype increases cardiovascular risk through many pathophysiological mechanisms [44], and this adds to the risk already intrinsic to MPNs.
These data suggest that the Body Mass Index (BMI) is a modifiable factor to be taken into consideration in the treatment of MPNs that could impact not only the quality of life but also the outcome. At the same time, it should be noted that ruxolitinib, a drug widely used in MPNs, has been associated with weight gain in patients due to its interactions with leptin [45]. These data should be taken into consideration, and obese patients might need their medication dosage adjusted or could benefit from more individualized nutrition advice. At the moment, the implications are not entirely clear, and ad hoc prospective studies are deemed necessary to understand the metabolic implications of ruxolitinib therapy and also the correct management of this patient setting.

2.4. Malnutrition

On the other hand, malnutrition, which is frequent particularly among patients with PMF, can have many implications.
The relationship between BMI and symptom burden in MPN patients is complex and requires careful interpretation. While patients with normal BMI generally experience the best quality of life, those with low BMI show increased constitutional symptoms [5].
Importantly, this relationship likely reflects involuntary weight loss associated with disease progression, cachexia, or malnutrition rather than intentional weight management. In patients with PV, progressive involuntary weight reduction (defined as a decrease of more than 10% in BMI) has been associated with worse overall survival [46]; cachexia in general is associated with disease progression and can be one of its first signals [47]. This finding does not contradict the potential benefits of targeted nutritional interventions aimed at metabolic optimization, as the mechanisms and clinical contexts differ substantially. Involuntary weight loss in MPN patients typically reflects disease-related catabolism and poor nutritional status, whereas structured dietary approaches focus on metabolic pathway modulation while maintaining adequate nutritional support and lean body mass.
Malnutrition also impacts the outcome due to its effect of compromising the immune function, thus increasing the risk of infections but also of treatment tolerance.
Although the pathogenetic mechanism may differ partly from that present in obese patients, inflammation also plays an important role in this case [48]. Many scores have been attempted to test the degree of malnutrition, among which many included serum albumin concentration and lymphocyte count, as in the Prognostic Nutritional Index (PNI). Lucijanic et al. have demonstrated that PMF patients with low serum albumin levels exhibit several concerning clinical features: increased fibrosis severity, markers of more aggressive disease (elevated LDH, peripheral blood blasts, and transfusion dependence), higher inflammatory markers like C-reactive protein (CRP), and ultimately, shorter survival times [49]. The improvement in body weight and albumin secondary to therapy with ruxolitinib could partly contribute to the survival advantage in patients treated with the JAK2 inhibitor; the proposed mechanism for this off-target effect of ruxolitinib is a reduction in JAK2-mediated phosphorylation of STAT3 in the arcuate nucleus of the murine hypothalamus in response to feeding or exogenous leptin [50].
According to Lucijanic M et al., lymphocyte counts are typically reduced in primary myelofibrosis patients, and incorporating these parameters into the PNI score creates an independent survival predictor relative to the DIPSS classification, particularly for those with the most diminished PNI values.
Performing the measurement of these two parameters is feasible and economical, and this information could help identify which patients face the highest risk and would benefit most from nutritional support [49].
In conclusion, this U-shaped relationship between BMI and the course of MPNs highlights the importance of monitoring the nutritional status of the patient and potentially incorporating nutritional and lifestyle interventions alongside pharmacological treatment for MPNs.

2.5. The Role of Microbiota

Many of the effects of food pass through a remodeling of the microbiota, which acts as an interface between the host and the diet. In MPNs, we know that inflammation plays a fundamental role; it is possible to hypothesize that, being a close link between microbiota and inflammation, the microbiota could contribute to the inflammatory trigger typical of MPNs. On the other hand, inflammation itself can alter the microbiota in a dysbiotic direction.
Few data are known to date. Oliver et al. demonstrated that MPN patients have a distinct microbiota composition compared to healthy individuals, characterized by lower levels of Phascolarctobacterium bacteria and reduced concentrations of short-chain fatty acids, particularly propionate. The reduction in Phascolarctobacterium is also characteristic of autoimmune diseases, where it plays a role in modulating inflammation [51]. In contrast, a different study comparing microbiota samples from 25 MPN patients and 23 healthy controls found no significant differences between these groups and similarly detected no microbiota distinctions between patients with high versus low symptom burden. The only notable finding was a 20% higher abundance of bacteria from the Prevotellaceae family in MPN patients; notably, patients receiving ruxolitinib therapy showed distinct microbiome profiles compared to those treated with hydroxyurea [52]. Recent research has shown that PV patients exhibit decreased alpha diversity in their gut microbiome and lower proportions of various Firmicutes bacterial taxa compared to healthy individuals; interestingly, patients treated with interferon had a microbiota composition more similar to that of healthy subjects, suggesting what has already been published, namely that IFN, as an anti-inflammatory agent, may be able to restore intestinal permeability [53,54]. Intestinal permeability does indeed appear altered in patients with PV, as demonstrated by the increase in circulating lipopolysaccharide (LPS) compared to healthy subjects [55].
Overall, the studies we have available are few and have not clarified to date the role that microbiota may have in the pathogenesis and perpetuation of the disease, as demonstrated for other cancers. However, we believe that these data, although preliminary, suggest that the role of the microbiota in MPNs should not be overlooked.
In general, we can affirm, based on the data present in the literature, that inflammation plays a key role in the pathogenesis of the disease, and a dietary approach aimed at reducing inflammation, with a role partly mediated by the microbiota, may have a beneficial role. Also paying attention to the nutritional status of the patient, the possible cardiovascular risk added by being overweight and diet, as well as the effect of the drugs used, must be taken into consideration.

3. The Role of Supplements in MPNs

The use of supplements is increasingly widespread and recognized among patients with lymphoma and myeloma, while data for MPNs are emerging; the results from the Nutrient Survey show that 72% of MPN respondents reported the use of supplements [56]. The most commonly used supplements by patients according to this survey are amino acid supplements, N-acetyl cysteine (NAC), Bach flower remedies, vitamin D, multivitamins, omega-3 fatty acids, calcium, turmeric, green tea, vitamin E, medical marijuana, and medicinal mushrooms. Comparable results also emerge from the SIMM study [9]. In most cases, patients do not report their use to physicians or, in any case, are not advised by physicians on which supplements to use. On the other hand, healthcare professionals do not have access to much evidence-based data, as most studies, as we will see, are based on preclinical models (cell and mouse models). A review of potentially beneficial supplements or those potentially harmful—for example, due to drug interactions—is therefore the purpose of this section.

3.1. Curcumin

In recent years, curcumin, a component of Curcuma Longa root, has shown its pleiotropic activity due to its ability to modulate many intracellular signal pathways.

3.1.1. Preclinical Studies

In MPNs, its action works through many mechanisms. The main one is the anti-inflammatory effect: as we have seen, inflammation plays a key role in the pathogenesis of MPNs, and curcumin has a well-demonstrated anti-inflammatory effect [57], contributing to reducing the cytokine burden. Curcumin can also induce apoptosis while simultaneously inhibiting anti-apoptotic proteins, such as Bcl2 and NFkB [58]. It has been demonstrated that in chronic myeloid leukemia, its anti-apoptotic action occurs through the upregulation of the PTEN oncogene, which is a target of miR-21, overexpressed in many cancer cells [59]. Its ability to also inhibit NFkB, a crucial factor in the activation of inflammatory, proliferative, and anti-apoptotic pathways, could play a key role [58]. Finally, it inhibits the JAK/STAT pathway, constitutively activated in MPNs [60,61].

3.1.2. Clinical Evidence

Despite promising preclinical results, clinical studies are lacking and necessary to understand the potential benefit of this substance in MPN patients and the possible interactions with JAK inhibitors and new classes of drugs. While data on pharmacokinetics and safety are available, as well as new high-availability formulations, the possible interaction with anticoagulant or anti-platelet drugs (almost always used in MPN patients) represents a difficult limitation to overcome in this patient setting; large-scale, randomized clinical trials are therefore necessary to define whether and how to use this valuable supplement. The NCT06063486 trial is currently ongoing with the aim of evaluating changes in inflammatory response and symptomatology in patients with clonal cytopenia of undetermined significance (CCUS), low-risk myelodysplastic syndrome (LR-MDS), and MPNs [62].

3.2. Vitamin D

Vitamin D deficiency is very common in the population and is associated with a higher incidence of cancer; in MPNs, lower vitamin D levels occur particularly in patients with PMF, and severe deficiency (levels below 10 ng/mL) occurs in patients with PMF and ET. However, the deficiency does not seem to correlate with clinical or laboratory variables, nor with OS or leukemia-free survival [63].

3.2.1. Preclinical Studies

Preclinical studies have provided conflicting evidence regarding vitamin D’s role in MPNs. There is limited evidence that vitamin D may interfere with the JAK2/STAT pathway [64]. However, murine models have demonstrated that vitamin D appears to stimulate bone marrow fibrosis through bone marrow macrophages, promoting the proliferation of collagen-producing fibroblasts and consequent reversible bone marrow fibrosis. Preclinical models lacking the vitamin D receptor, as well as vitamin D deprivation itself, are not associated with myelofibrosis, which would be independent of TGFβ1 or megakaryocytes [65]. These findings contradict previous observations regarding the presence of bone marrow fibrosis in children with rickets [66], which was reversible after vitamin D supplementation, as well as the proven ability of vitamin D to reduce circulating TGF β1 [67].

3.2.2. Clinical Evidence

Clinical data on vitamin D in MPNs remain limited. Vitamin D could be beneficial due to its protective role against thrombosis and may therefore reduce cardiovascular risk in MPN patients [68]. Additionally, there is preliminary evidence that vitamin D can reduce platelet count in ET patients [69]. Although supplementing vitamin D in deficient patients might be recommendable, the evidence that vitamin D can stimulate bone marrow fibrosis makes the correct therapeutic approach controversial.
More data are needed to better understand vitamin D supplementation in MPNs, and an investigation of vitamin D receptor antagonists as anti-fibrotic agents could be considered.

3.3. Omega-3 Fatty Acids

3.3.1. Preclinical Studies

Omega-3 fatty acids, particularly eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), have well-established anti-inflammatory effects that contribute to reducing the cytokine burden and resolving inflammation through specialized pro-resolving mediators (SPMs) [70]. Due to their anti-platelet aggregation effect, they can attenuate the hypercoagulable state characteristic of MPNs [71]. Additionally, preclinical studies have demonstrated that omega-3 fatty acids are able to reduce myeloid progenitors and increase cell differentiation [72].

3.3.2. Clinical Evidence

Although there are no specific clinical studies on omega-3 fatty acids in MPNs, recent clinical evidence has addressed the safety concerns regarding their use. Claims regarding a possible enhancement of anti-platelet action have recently been refuted by a large meta-analysis [73]. Therefore, the use of omega-3 fatty acids could be considered due to their established anti-inflammatory effect and safety profile.

3.4. N-Acetylcysteine (NAC)

NAC is a sulfhydryl-containing compound with notable antioxidant properties that demonstrates several mechanisms of potential benefit in MPNs.

3.4.1. Preclinical Studies

It has a potent antioxidant effect as a precursor of glutathione, which is particularly relevant, since elevated reactive oxygen species (ROS) levels have been demonstrated in MPNs, amplified by JAK2 mutations [74]. NAC is also characterized by anti-inflammatory properties and inhibitory effects on NFkB [75,76]. Additionally, preclinical studies have shown anti-fibrotic action in other organs, such as liver and lung, although studies specifically on bone marrow are lacking [77].

3.4.2. Clinical Evidence

Currently, there are limited clinical data specifically addressing NAC use in MPN patients. Orally soluble formulations with enhanced bioavailability and intravenous formulations are now available. While formal clinical trials in MPN patients are lacking, preliminary clinical observations suggest good tolerability, although systematic evaluation remains necessary.

3.5. Artemisinin

Artemisinin is a sesquiterpene lactone derived from the plant Artemisia annua, used for centuries in traditional Chinese medicine.

3.5.1. Preclinical Studies

Preclinical evidence suggests that it may have specific anti-fibrotic action in addition to its anti-inflammatory and proposed anti-cancer properties. The potential mechanisms through which it would exert its action in MPNs include JAK2/STAT3 downregulation [78] and formation of iron-dependent free radicals: artemisinin can cause cell death through the generation of reactive oxygen species from iron, a molecule in which cancer cells are particularly rich, making them more susceptible to so-called ferroptosis [79].
A preclinical study on zebrafish demonstrated that artemisinin can block erythroid production in cells carrying the JAK2 AV581F mutation, the zebrafish counterpart of the JAK2 V617F mutation in human PV cells [80]. Anti-fibrotic action has been demonstrated on pulmonary, renal, and hepatic tissue and is mediated by inhibition of TGFβ, inflammation, epithelial–mesenchymal transition (EMT), and myofibroblast activation. Since the anti-fibrotic mechanism seems to be common to all tissues, this could also occur in the bone marrow, although specific studies still need to be conducted [81].

3.5.2. Clinical Evidence

Artemisinin has demonstrated a favorable safety profile in malaria therapy. Although preliminary clinical observations in MPN patients suggest acceptable tolerability and potential stabilization of blood counts, these findings require validation through controlled clinical studies.

3.6. Vitamin C

In a real-world analysis, vitamin C deficiency was demonstrated in 17% of patients with myeloid hematological diseases; the deficiency was more frequent in young people, in association with the AXL1 mutation or with acute myeloid leukemia [82].

Clinical Evidence

Vitamin C appears to particularly have a role in myelodysplastic syndromes, acute myeloid leukemias, and clonal cytopenias due to its epigenetic action through the modulation of TET2 [83] and synergy with azacitidine or decitabine [84,85].
In myeloproliferative diseases, however, its role is controversial. TET2 mutations have also been found in ET, PV, and PMF, although their prognostic impact appears limited [86].
Since vitamin C could potentially increase iron absorption and exacerbate red blood cell production in PV, it is typically not recommended for PV patients.
The observation that intravenous vitamin C administered for many consecutive days causes an alteration in thromboelastogram and platelet function raises further caution in using vitamin C in ET patients as well [87].
Overall, despite the positive role in some myeloid neoplasms, its role in myeloproliferative diseases is debated, and without consistent data based on evidence at the moment, its use in these pathologies is not recommended but requires further study.

3.7. Quercetin

Quercetin is a flavonoid compound found in many vegetables, belonging to the class of polyphenols; it is studied for its potential health benefits, among which the main ones are anti-inflammatory and antioxidant.

3.7.1. Preclinical Studies

Quercetin has been shown to synergize with BET inhibitors in vitro, which is particularly relevant, since MPNs recognize inflammation as a fundamental trigger, and BET pathway inhibitors are under investigation for approval [88].
Quercetin also has the ability to promote apoptosis and autophagy through the modulation of PI3K/Akt/mTOR, Wnt/β-catenin, and MAPK/ERK1/2 pathways [89] and can inhibit the JAK2 pathway [90]. Through anti-inflammatory action, inhibition of TGFβ, and modulation of myofibroblast formation, quercetin presents itself as an anti-fibrotic agent that could potentially be effective in myelofibrosis, although preclinical and clinical studies confirming this hypothesis are currently lacking [91].

3.7.2. Clinical Evidence

A clinical study is currently underway that involves the association of dasatinib and quercetin in idiopathic pulmonary fibrosis, precisely by virtue of their senolytic and anti-fibrotic action [92].
Based on these mechanisms of action and considering the availability of liposomal formulations with greater bioavailability, quercetin appears as a potential therapeutic tool in MPN patients requiring further study.

4. Acupuncture in MPNs

Acupuncture has been explored as a supportive therapy in MPNs, with patient surveys indicating interest in its use for symptom relief. The SIMM Study-2, a survey-based study, highlighted that patients with MPN actively seek acupuncture treatments to complement conventional care and enhance their QoL [9]. While data on acupuncture in MPN remain limited, case reports and broader IO research support its potential benefits.
Pruritus, a debilitating symptom in PV, is often resistant to conventional treatment. Although not specifically studied in MPN patients, acupuncture has been shown to improve this sensation in various diseases via inhibition of peripheral and central transmission of itching [93]. A case report documented notable symptom relief in a patient with PV following acupuncture treatment, highlighting marked improvements in itching, fatigue, bone pain, and headache. These findings suggest a potential role of acupuncture in modulating neuroimmune responses and alleviating inflammation [94]. Similarly, erythromelalgia, a painful microvascular disorder seen in ET and PV, has shown responsiveness to acupuncture and Kampo medicine in a case report, supporting its vasoregulatory and analgesic effect [95]. Fatigue, another hallmark symptom of MPN, has been a focus of integrative interventions, including acupuncture, in both solid and hematologic malignancies, although not specifically studied in MPN patients [96,97]. The proposed mechanisms of action involve anti-inflammatory effect and neuromodulation [98]. Additionally, acupuncture has been studied in related hematologic malignancies and bone marrow transplanted patients, demonstrating its efficacy in improving nausea and vomiting, sleep disturbances, QoL, and overall well-being [98,99], reinforcing its potential in MPN management.
While acupuncture is generally safe, careful consideration is required in MPN patients, particularly those with thrombocytopenia or an elevated risk of bleeding. The risk of infection is low, but immunocompromised patients should receive treatment from trained professionals under strict aseptic conditions [100,101]. Evidence from hematologic malignancies suggests that acupuncture can be safely administered even in patients with low platelet counts (below 20 × 109/L), reinforcing its feasibility in patients with MPN [102]. In conclusion, acupuncture presents a promising adjunctive therapy for MPN-related symptoms, addressing unmet needs in symptom management and patient well-being. Further clinical studies are warranted to establish standardized protocols and expand its evidence base in this patient population.

5. Physical Activity in MPNs

Most exercise-focused research in hematologic cancers has concentrated on leukemia, lymphoma, and multiple myeloma, though emerging data now point to benefits for MPN patients as well [103]. Physical activity has been shown to mitigate a range of cancer-related adverse effects, including fatigue, cognitive changes, psychosocial distress, sexual dysfunction, and overall declines in the quality of life [104]. Furthermore, individuals who maintain regular physical activity have a reduced risk of several malignancies, such as colorectal, liver, esophageal, lung, leukemia, and melanoma [105]. In addition, exercise has been associated with decreased cancer-specific and all-cause mortality, particularly in those with breast and colorectal cancer [106].
Mechanistically, exercise may enhance the immune function, potentially explaining its anti-cancer effects. For instance, the mobilization of natural killer (NK) cells and IL-6 release during physical activity has been linked to tumor suppression in preclinical models [107,108,109]. Moreover, exercise has been shown to modulate insulin-related growth factors that may play a role in cancer progression, especially in breast cancer [110]. Other theorized mechanisms include improved treatment tolerance leading to greater therapy completion, as well as beneficial changes in the epigenome [111,112].
In the context of MPNs, exercise may be especially important due to the heightened risk of thrombotic events, with cardiovascular fitness potentially improved through enhanced fibrinolysis [113]. Sedentary behavior in this population has been linked to worse fatigue and diminished quality of life [114,115]. A large, international, multicenter patient reported survey suggested that structured aerobic and resistance training was associated with reduced symptom burden (p = 0.01) and lower rates of depression (p = 0.006), though other studies have reported mixed results [116]. One 12-week exercise trial in MPNs found no significant improvements in fatigue or quality of life, possibly due to differences in adherence, patient characteristics, or trial design [113]. Nonetheless, improved physical strength (p = 0.01, p < 0.001) and increased VO2 max (p = 0.01) were observed, highlighting the feasibility and impact of exercise in this population [117].
The optimal type, frequency, and intensity of exercise for MPNs remains unclear. The current guidance often draws from NCCN recommendations for cancer survivors, which advocate for 150 min of moderate aerobic activity and strength training weekly, spread over at least two days [118]. Among polycythemia vera patients, preferences tend to lean toward individualized, outdoor-based programs performed twice weekly for 45–60 min [119]. Exercise plans may need adjustment for individuals with splenomegaly, bleeding risks, or pain, such as avoiding high-impact sports. Barriers to participation are common, and addressing these through cognitive support and personalized exercise plans may improve engagement [119,120].

6. Mind–Body Therapies in MPNs

Mind–body therapies (MBTs) integrate mindful awareness, physical movement, and breathing techniques to support psychological and physiological health [121]. Approaches such as yoga, mindfulness/meditation, tai chi, and qigong have demonstrated benefits in alleviating cancer-related symptoms like anxiety, depression, pain, and sleep disturbances [122]. These effects are believed to occur through multiple pathways, including dampening systemic inflammation, as evidenced by reductions in CRP and IL-6, and transcriptional shifts that downregulate NF-kB activity while enhancing glucocorticoid receptor signaling [123]. MBTs may also influence central and autonomic nervous system regulation, fostering self-compassion and reducing maladaptive thought patterns [124].
Yoga, for example, has been shown to improve psychosocial outcomes in patients with breast cancer and lymphoma [125]. In the MPN setting, a 12-week online yoga program (60 min per week) led to improvements in sleep, symptom burden, mood, and pain [126]. Patients reported benefits in circulation, respiratory function, dietary habits, and enjoyment [127,128]. These findings are supported by high levels of satisfaction, adherence, and follow-up [129]. Biologically, yoga reduced plasma TNF levels in MPNs, although IL-6 remained unchanged, with benefits more prominent in individuals with higher BMI [130]. Some participants experienced discomfort, such as spleen irritation, but these were manageable through modified poses [131]. Barriers to participation can often be mitigated by remote formats and low-dose interventions. To date, recommendations regarding yoga style, frequency, and intensity remain vague, though Hatha and Vinyasa practices of mild to moderate intensity are commonly used. Notably, sessions exceeding 60 min weekly did not yield additional benefits, highlighting the need for larger, controlled trials [130,131].
Mindfulness, defined as an intentional, non-judgmental awareness of the present moment, has also gained traction as an intervention in MPNs [132]. Mobile apps like Calm and My Wellness Coach (MWC) have demonstrated feasibility and patient satisfaction, with users often recommending these tools to peers [133,134]. Evidence suggests that mindfulness apps can reduce anxiety and depressive symptoms, particularly among patients with pre-existing mental health challenges [133,134]. MWC also appeared to improve symptoms such as dizziness, night sweats, sexual dysfunction, bone pain, cognitive difficulties, and sedentary behavior. Participants noted enhanced tranquility and overall well-being, though improvements in sleep were limited, perhaps due to shorter intervention durations (4–12 weeks) [133,134].
Technical limitations like poor internet access, annoying app notifications, and tech literacy challenges, especially in older populations, were noted [135,136]. Optimal dosage remains uncertain, as no significant difference in outcomes was observed between users engaging in 10 versus 30 min of mindfulness per session [137].

7. Other Integrative Approaches for MPNs

Integrative therapies, such as massage, music therapy, art therapy, Aryuveda, healing touch, and other therapies, are increasingly recognized for their potential to alleviate symptom burden and enhance QoL in patients with cancer. These integrative modalities may offer supportive benefit by reducing psychological sequelae of the MPNs while concurrently treating the physical contributors of symptom burden associated with MPN. Despite promising preliminary evidence in other cancer populations, there is a critical gap in rigorous research evaluating the efficacy, feasibility, and mechanistic impact of these therapies in MPNs. Expanding clinical trials and implementation studies in this area could inform integrative care models and help optimize patient-centered outcomes in the MPN population.

8. Discussion

In this review, we attempted to collect all the available literature data of scientific value on the various tools of integrative medicine and their potential role in MPNs. The current landscape of integrative medicine in MPN management is characterized by significant research gaps that must be systematically addressed to advance clinical practice. The predominance of preclinical investigations over clinical studies represents the most critical limitation in this field. This evidence deficit stems from the relatively recent recognition of integrative approaches in hematology and the inherent complexity of conducting controlled trials in rare disease populations.
The scarcity of disease-specific clinical evidence for MPN patients represents a fundamental challenge. While general principles from oncology and other chronic conditions may be applicable, the unique pathophysiology of MPNs—particularly their inflammatory nature and JAK/STAT pathway involvement—demands targeted research approaches. The current gaps include the lack of standardized protocols for dietary interventions, insufficient safety data for supplement use in the context of standard therapies, and limited understanding of optimal timing and dosing for integrative interventions.
Despite these limitations, integrative medicine offers significant potential for MPN management. The inflammatory nature of MPNs provides a strong rationale for anti-inflammatory dietary interventions and lifestyle modifications that may address both symptoms and disease progression. Moreover, integrative approaches represent a valuable opportunity for patients to actively participate in their care journey, potentially enhancing their sense of empowerment and overall well-being while addressing the multidimensional aspects of their condition. However, any recommendations in this domain must adhere to the fundamental principle of “Primum non nocere” (First do no harm), ensuring that integrative interventions serve as valuable adjuncts to established evidence-based treatments.
The current research priorities center on understanding the anti-inflammatory potential of dietary modifications, particularly the Mediterranean diet, given the established inflammatory trigger in MPNs. The investigation of specific compounds with JAK/STAT pathway modulatory effects—including artemisinin, curcumin, and NAC—represents another critical area. Additionally, symptom management through non-pharmacological approaches, such as acupuncture and mindfulness interventions, shows promise for improving quality of life outcomes.
The advancement of integrative medicine in MPN care requires collaborative efforts between hematologists, integrative medicine specialists, and researchers to establish a robust evidence base that ensures patient safety while maximizing therapeutic potential. This coordinated approach will enable the development of comprehensive treatment strategies that address both the biological and psychosocial aspects of MPN management. Table 1 summarizes the potential integrative approaches and research directions in the MPN setting.

9. Conclusions

In conclusion, integrative therapies hold significant promise for improving the symptom burden and quality of life among patients with MPNs. However, rigorous research is urgently needed to evaluate the effects of key integrative interventions, such as nutrition, physical activity, dietary supplements, and mind–body practices, on both subjective and objective outcomes. Beyond symptom control, future studies must examine how these interventions influence critical clinical endpoints, including thrombosis, leukemic transformation, and overall survival. To fully realize the potential of integrative care in MPNs, robust translational research is essential. Such efforts should investigate the biological underpinnings of integrative interventions, including their effects on mutational allele burdens, clonal evolution, cardiovascular risk markers, inflammatory pathways, immune phenotypes, and the microbiome. Through multidisciplinary collaboration and thoughtfully designed trials, we can bridge the gap between integrative approaches and precision medicine, advancing whole-person care for those living with MPNs.

Author Contributions

Conceptualization, F.A. and M.B.; writing—original draft preparation, F.A., I.L.Y., and K.G.; writing—review and editing, F.A., I.L.Y., and K.G.; supervision, M.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

This review article did not generate any new data.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
BETBromodomain and Extra-Terminal Motif
BCL2B-Cell Lymphoma
BMIBody Mass Index
CIMComplementary and Integrative Medicine
CRPC-Reactive Protein
DHADocosahexaenoic Acid
DIPSSDynamic International Prognostic Scoring System
EPAEicosapentaenoic Acid
EpoErythropoietin
ETEssential Thrombocythemia
GLUT1Glucose Transporter 1
IGF1Insulin-Like Growth Factor 1
IHIntegrative Hematology
IL-1βInterleukin-1β
IL6Interleukin 6
IL8Interleukin 8
IOIntegrative Oncology
JAKJanus Chinasi
MAPKMitogen-Activated Protein Kinase
MGUSMonoclonal Gammopathy of Undetermined Significance
MMMultiple Myeloma
MPNsMyeloproliferative Neoplasms
NACN-Acetyl Cysteine
OSOverall Survival
PFKFB36-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3
PFSProgression-Free Survival
PMFPrimary Myelofibrosis
PNIPrognostic Nutritional Index
PTENPhosphatase and Tensin Homolog
PVPolicytemia Vera
QoLQuality of Life
SIMMSurvey of Integrative Medicine in Myeloproliferative Neoplasms
STATSignal Transducer and Activator of Transcription
TET2Tet Methylcytosine Dioxygenase 2
TGFTransforming Growth Factor
TNF αTumor Necrosis Factor-α
USDAUS Dietary Guidelines for Americans
VEGFVascular Endothelium Growth Factor

References

  1. Tefferi, A.; Pardanani, A. Myeloproliferative Neoplasms: A Contemporary Review. JAMA Oncol. 2015, 1, 97–105. [Google Scholar] [CrossRef] [PubMed]
  2. Arber, D.A.; Orazi, A.; Hasserjian, R.; Thiele, J.; Borowitz, M.J.; Le Beau, M.M.; Bloomfield, C.D.; Cazzola, M.; Vardiman, J.W. The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood 2016, 127, 2391–2405. [Google Scholar] [CrossRef] [PubMed]
  3. Verstovsek, S.; Mesa, R.A.; Gotlib, J.; Levy, R.S.; Gupta, V.; DiPersio, J.F.; Catalano, J.V.; Deininger, M.; Miller, C.; Silver, R.T.; et al. A double-blind, placebo-controlled trial of ruxolitinib for myelofibrosis. N. Engl. J. Med. 2012, 366, 799–807. [Google Scholar] [CrossRef] [PubMed]
  4. Scherber, R.; Dueck, A.C.; Johansson, P.; Barbui, T.; Barosi, G.; Vannucchi, A.M.; Passamonti, F.; Andreasson, B.; Ferarri, M.L.; Rambaldi, A.; et al. The Myeloproliferative Neoplasm Symptom Assessment Form (MPN-SAF): International prospective validation and reliability trial. Blood 2011, 118, 401–408. [Google Scholar] [CrossRef] [PubMed]
  5. Mesa, R.A.; Niblack, J.; Wadleigh, M.; Verstovsek, S.; Camoriano, J.; Barnes, S.; Tan, A.D.; Atherton, P.J.; Sloan, J.A.; Tefferi, A. The burden of fatigue and quality of life in myeloproliferative disorders: An International Internet-Based Survey. Cancer 2007, 109, 68–76. [Google Scholar] [CrossRef] [PubMed]
  6. Geyer, H.L.; Scherber, R.M.; Dueck, A.C.; Kiladjian, J.J.; Xiao, Z.; Slot, S.; Zweegman, S.; Sackmann, F.; Fuentes, A.K.; Hernández-Maraver, D.; et al. Distinct clustering of symptom burden among myeloproliferative neoplasm patients: Retrospective assessment in 1470 patients. Blood 2014, 123, 3803–3810. [Google Scholar] [CrossRef] [PubMed]
  7. Mesa, R.A.; Scherber, R.M.; Geyer, H.L. Reducing symptom burden in patients with myeloproliferative neoplasms in the era of Janus kinase inhibitors. Leuk. Lymphoma 2015, 56, 1989–1999. [Google Scholar] [CrossRef] [PubMed]
  8. Connor, K.M.; Davidson, J.R. Development of a new resilience scale: The Connor-Davidson Resilience Scale (CD-RISC). Depress. Anxiety 2003, 18, 76–82. [Google Scholar] [CrossRef] [PubMed]
  9. Gowin, K.; Langlais, B.T.; Kosiorek, H.E.; Dueck, A.; Millstine, D.; Huberty, J.; Eckert, R.; Mesa, R.A. The SIMM study: Survey of integrative medicine in myeloproliferative neoplasms. Cancer Med. 2020, 9, 9445–9453. [Google Scholar] [CrossRef] [PubMed]
  10. Yun, H.; Sun, L.; Mao, J.J. Growth of integrative medicine at leading cancer centers between 2009 and 2016: A systematic analysis of NCI-designated comprehensive cancer center websites. J. Natl. Cancer Inst. Monogr. 2017, 2017, lgx004. [Google Scholar] [CrossRef] [PubMed]
  11. Witt, C.M.; Balneaves, L.G.; Cardoso, M.J.; Cohen, L.; Greenlee, H.; Johnstone, P.; Kücük, Ö.; Mailman, J.; Mao, J.J. A comprehensive definition for integrative oncology. J. Natl. Cancer Inst. Monogr. 2017, 2017, lgx012. [Google Scholar] [CrossRef] [PubMed]
  12. Andreazzoli, F.; Bonucci, M. Integrative Hematology: State of the Art. Int. J. Mol. Sci. 2023, 24, 1732. [Google Scholar] [CrossRef] [PubMed]
  13. Surapaneni, P.; Scherber, R.M. Integrative Approaches to Managing Myeloproliferative Neoplasms: The Role of Nutrition, Exercise, and Psychological Interventions. Curr. Hematol. Malig. Rep. 2019, 14, 164–170. [Google Scholar] [CrossRef] [PubMed]
  14. Singh, S.; Peshin, S.; Larsen, A.; Gowin, K. Optimizing care: Integrative oncology in myeloproliferative neoplasm. Curr. Oncol. Rep. 2024, 26, 1135–1145. [Google Scholar] [CrossRef] [PubMed]
  15. WCRF Third Report. Available online: https://www.wcrf.org/wp-content/uploads/2024/11/Summary-of-Third-Expert-Report-2018.pdf (accessed on 30 April 2025).
  16. Shah, U.A.; Parikh, R.; Castro, F.; Bellone, M.; Lesokhin, A.M. Dietary and microbiome evidence in multiple myeloma and other plasma cell disorders. Leukemia 2023, 37, 964–980. [Google Scholar] [CrossRef] [PubMed]
  17. Malik, M.A.; Sweeney, N.W.; Jafri, M.; Derkach, A.; Chmielewski, C.; Adintori, P.A.; Mailankody, S.; Korde, N.; Tan, C.R.; Hassoun, H.; et al. Nutrition perceptions, needs and practices among patients with plasma cell disorders. Blood Cancer J. 2022, 12, 70. [Google Scholar] [CrossRef] [PubMed]
  18. Geyer, H.L.; Dueck, A.C.; Scherber, R.M.; Mesa, R.A. Impact of Inflammation on Myeloproliferative Neoplasm Symptom Development. Mediat. Inflamm. 2015, 2015, 284706. [Google Scholar] [CrossRef] [PubMed]
  19. Fleischman, A.G. Inflammation as a Driver of Clonal Evolution in Myeloproliferative Neoplasm. Mediat. Inflamm. 2015, 2015, 606819. [Google Scholar] [CrossRef] [PubMed]
  20. Longhitano, L.; Li Volti, G.; Giallongo, C.; Spampinato, M.; Barbagallo, I.; Di Rosa, M.; Romano, A.; Avola, R.; Tibullo, D.; Palumbo, G.A. The Role of Inflammation and Inflammasome in Myeloproliferative Disease. J. Clin. Med. 2020, 9, 2334. [Google Scholar] [CrossRef] [PubMed]
  21. Mendez Luque, L.F.; Avelar-Barragan, J.; Nguyen, H.; Nguyen, J.; Soyfer, E.M.; Liu, J.; Chen, J.H.; Mehrotra, N.; Kosiorek, H.E.; Dueck, A.; et al. The NUTRIENT Trial (NUTRitional Intervention among myEloproliferative Neoplasms): Feasibility Phase. medRxiv 2023, 2023.05.09.23289740. [Google Scholar] [CrossRef] [PubMed]
  22. Chen, E.C.; Johnston, H.; Patel, A.A. Targeted Therapy for MPNs: Going Beyond JAK Inhibitors. Curr. Hematol. Malig. Rep. 2023, 18, 41–55. [Google Scholar] [CrossRef] [PubMed]
  23. Tosti, V.; Bertozzi, B.; Fontana, L. Health Benefits of the Mediterranean Diet: Metabolic and Molecular Mechanisms. J. Gerontol. A Biol. Sci. Med. Sci. 2018, 73, 318–326. [Google Scholar] [CrossRef] [PubMed]
  24. Estruch, R.; Ros, E.; Salas-Salvadó, J.; Covas, M.I.; Corella, D.; Arós, F.; Gómez-Gracia, E.; Ruiz-Gutiérrez, V.; Fiol, M.; Lapetra, J.; et al. Primary Prevention of Cardiovascular Disease with a Mediterranean Diet Supplemented with Extra-Virgin Olive Oil or Nuts. N. Engl. J. Med. 2018, 378, e34. [Google Scholar] [CrossRef] [PubMed]
  25. Andreazzoli, F.; Fiorito, A.; Bonucci, M. ARTOI Nutritional Approach in the Hematological Patient: Is there a Rationale? J. Biomed. Res. Rev. 2019, 2, 50–55. [Google Scholar]
  26. Hsieh, H.H.; Yao, H.; Ma, Y.; Zhang, Y.; Xiao, X.; Stephens, H.; Wajahat, N.; Chung, S.S.; Xu, L.; Xu, J.; et al. Epo-IGF1R cross talk expands stress-specific progenitors in regenerative erythropoiesis and myeloproliferative neoplasm. Blood 2022, 140, 2371–2384. [Google Scholar] [CrossRef] [PubMed]
  27. Staerk, J.; Kallin, A.; Royer, Y.; Diaconu, C.C.; Dusa, A.; Demoulin, J.B.; Vainchenker, W.; Constantinescu, S.N. JAK2, the JAK2 V617F mutant and cytokine receptors. Pathol. Biol. 2007, 55, 88–91. [Google Scholar] [CrossRef] [PubMed]
  28. Melnik, B.C.; John, S.M.; Schmitz, G. Over-stimulation of insulin/IGF-1 signaling by Western diet may promote diseases of civilization: Lessons learnt from Laron syndrome. Nutr. Metab. 2011, 8, 41. [Google Scholar] [CrossRef] [PubMed]
  29. Yeh, H.C.; Maruthur, N.M.; Wang, N.Y.; Jerome, G.J.; Dalcin, A.T.; Tseng, E.; White, K.; Miller, E.R.; Juraschek, S.P.; Mueller, N.T.; et al. Effects of Behavioral Weight Loss and Metformin on IGFs in Cancer Survivors: A Randomized Trial. J. Clin. Endocrinol. Metab. 2021, 106, e4179–e4191. [Google Scholar] [CrossRef] [PubMed]
  30. Rahmani, J.; Montesanto, A.; Giovannucci, E.; Zand, H.; Barati, M.; Kopchick, J.J.; Mirisola, M.G.; Lagani, V.; Bawadi, H.; Vardavas, R.; et al. Association between IGF-1 levels ranges and all-cause mortality: A meta-analysis. Aging Cell 2022, 21, e13540. [Google Scholar] [CrossRef] [PubMed]
  31. Hosseinzadeh, Z.; Bhavsar, S.K.; Shojaiefard, M.; Saxena, A.; Merches, K.; Sopjani, M.; Alesutan, I.; Lang, F. Stimulation of the glucose carrier SGLT1 by JAK2. Biochem. Biophys. Res. Commun. 2011, 408, 208–213. [Google Scholar] [CrossRef] [PubMed]
  32. Rao, T.N.; Hansen, N.; Hilfiker, J.; Rai, S.; Majewska, J.M.; Leković, D.; Gezer, D.; Andina, N.; Galli, S.; Cassel, T.; et al. JAK2-mutant hematopoietic cells display metabolic alterations that can be targeted to treat myeloproliferative neoplasms. Blood 2019, 134, 1832–1846. [Google Scholar] [PubMed]
  33. Reddy, M.M.; Fernandes, M.S.; Deshpande, A.; Weisberg, E.; Inguilizian, H.V.; Abdel-Wahab, O.; Kung, A.L.; Levine, R.L.; Griffin, J.D.; Sattler, M. The JAK2V617F oncogene requires expression of inducible phosphofructokinase/fructose-bisphosphatase 3 for cell growth and increased metabolic activity. Leukemia 2012, 26, 481–489. [Google Scholar] [CrossRef] [PubMed]
  34. Mendez Luque, L.F.; Blackmon, A.L.; Ramanathan, G.; Fleischman, A.G. Key Role of Inflammation in Myeloproliferative Neoplasms: Instigator of Disease Initiation, Progression. and Symptoms. Curr. Hematol. Malig. Rep. 2019, 14, 145–153. [Google Scholar] [CrossRef] [PubMed]
  35. Duncombe, A.S.; Anderson, L.A.; James, G.; de Vocht, F.; Fritschi, L.; Mesa, R.; Clarke, M.; McMullin, M.F. Modifiable Lifestyle and Medical Risk Factors Associated With Myeloproliferative Neoplasms. Hemasphere 2020, 4, e327. [Google Scholar] [CrossRef] [PubMed]
  36. Leiba, A.; Duek, A.; Afek, A.; Derazne, E.; Leiba, M. Obesity and related risk of myeloproliferative neoplasms among israeli adolescents. Obesity 2017, 25, 1187–1190. [Google Scholar] [CrossRef] [PubMed]
  37. Sun, S.; Ji, Y.; Kersten, S.; Qi, L. Mechanisms of Inflammatory Responses in Obese Adipose Tissue. Annu. Rev. Nutr. 2012, 32, 261–286. [Google Scholar] [CrossRef] [PubMed]
  38. Tefferi, A.; Vaidya, R.; Caramazza, D.; Finke, C.; Lasho, T.; Pardanani, A. Circulating interleukin (IL)-8, IL-2R, IL-12, and IL-15 levels are independently prognostic in primary myelofibrosis: A comprehensive cytokine profiling study. J. Clin. Oncol. 2011, 29, 1356–1363. [Google Scholar] [CrossRef] [PubMed]
  39. Christensen, S.F.; Scherber, R.M.; Brochmann, N.; Goros, M.; Gelfond, J.; Andersen, C.L.; Flachs, E.M.; Mesa, R. Body Mass Index and Total Symptom Burden in Myeloproliferative Neoplasms Discovery of a U-shaped Association. Cancers 2020, 12, 2202. [Google Scholar] [CrossRef] [PubMed]
  40. Moon, Y.S.; Kim, D.H.; Song, D.K. Serum tumor necrosis factor-α levels and components of the metabolic syndrome in obese adolescents. Metabolism 2004, 53, 863–867. [Google Scholar] [CrossRef] [PubMed]
  41. Pourcelot, E.; Trocme, C.; Mondet, J. Cytokine profiles in polycythemia vera and essential thrombocythemia patients: Clinical implications. Exp. Hematol. 2019, 42, 360–368. [Google Scholar] [CrossRef] [PubMed]
  42. Bowers, E.; Singer, K. Obesity-induced inflammation: The impact of the hematopoietic stem cell niche. JCI Insight 2021, 6, e145295. [Google Scholar] [CrossRef] [PubMed]
  43. Nam, S.Y.; Lee, E.J.; Kim, K.R.; Cha, B.S.; Song, Y.D.; Lim, S.K.; Lee, H.C.; Huh, K.B. Effect of obesity on total and free insulin-like growth factor (IGF)-1, and their relationship to IGF-binding protein (BP)-1, IGFBP-2, IGFBP-3, insulin, and growth hormone. Int. J. Obes. Relat. Metab. Disord. 1997, 21, 355–359. [Google Scholar] [CrossRef] [PubMed]
  44. Gallo, G.; Desideri, G.; Savoia, C. Update on Obesity and Cardiovascular Risk: From Pathophysiology to Clinical Management. Nutrients 2024, 16, 2781. [Google Scholar] [CrossRef] [PubMed]
  45. Mollé, N.; Krichevsky, S.; Kermani, P.; Silver, R.T.; Ritchie, E.; Scandura, J.M. Ruxolitinib can cause weight gain by blocking leptin signaling in the brain via JAK2/STAT3. Blood 2020, 135, 1062–1066. [Google Scholar] [CrossRef] [PubMed]
  46. Jang, A.; Hamad, H.; Yellapragada, S.V.; Sosa, I.R.; Rivero, G.A. Weight Loss Predicts Inferior Outcome in Polycythemia Vera Patients. Blood 2020, 136 (Suppl. 1), 22–23. [Google Scholar] [CrossRef]
  47. Tefferi, A.; Nicolosi, M.; Penna, D.; Mudireddy, M.; Szuber, N.; Lasho, T.L.; Hanson, C.A.; Ketterling, R.P.; Gangat, N.; Pardanani, A.D. Development of a prognostically relevant cachexia index in primary myelofibrosis using serum albumin and cholesterol levels. Blood Adv. 2018, 2, 1980–1984. [Google Scholar] [CrossRef] [PubMed]
  48. Fearon, K.C.H.; Glass, D.J.; Guttridge, D.C. Cancer cachexia: Mediators, signaling and metabolic pathways. Cell Metab. 2012, 16, 153–166. [Google Scholar] [CrossRef] [PubMed]
  49. Lucijanic, M.; Veletic, I.; Rahelic, D.; Pejsa, V.; Cicic, D.; Skelin, M.; Livun, A.; Tupek, K.M.; Stoos-Veic, T.; Lucijanic, T.; et al. Assessing serum albumin concentration, lymphocyte count and prognostic nutritional index might improve prognostication in patients with myelofibrosis. Wien. Klin. Wochenschr. 2018, 130, 126–133. [Google Scholar] [CrossRef] [PubMed]
  50. Greenfield, G.; McMullin, M.F. Ruxolitinib: Gaining more than intended. Blood 2020, 135, 983–984. [Google Scholar] [CrossRef] [PubMed]
  51. Oliver, A.; El Alaoui, K.; Haunschild, C.; Avelar-Barragan, J.; Mendez Luque, L.F.; Whiteson, K.; Fleischman, A.G. Fecal Microbial Community Composition in Myeloproliferative Neoplasm Patients Is Associated with an Inflammatory State. Microbiol. Spectr. 2022, 10, e0003222. [Google Scholar] [CrossRef] [PubMed]
  52. Elalaoui, K.; Weihe, C.; Oliver, A.; Craver, B.; Lai, H.Y.; Brooks, S.B.; Kim, D.; Martiny, J.; Whiteson, K.; Fleischman, A. Investigating the Role of the Gut Microbiome in the Inflammatory State of Myeloproliferative Neoplasms. Blood 2018, 132 (Suppl. 1), 3051. [Google Scholar] [CrossRef]
  53. Eickhardt-Dalbøge, C.S.; Ingham, A.C.; Andersen, L.O.; Nielsen, H.V.; Fuursted, K.; Stensvold, C.R.; Larsen, M.K.; Kjær, L.; Christensen, S.F.; Knudsen, T.A.; et al. The gut microbiota in patients with polycythemia vera is distinct from that of healthy controls and varies by treatment. Blood Adv. 2023, 7, 3326–3337. [Google Scholar] [CrossRef] [PubMed]
  54. Kotredes, K.P.; Thomas, B.; Gamero, A.M. The Protective Role of Type I Interferons in the Gastrointestinal Tract. Front Immunol. 2017, 8, 410. [Google Scholar] [CrossRef] [PubMed]
  55. Barone, M.; Barone, M.; Ricci, F.; Auteri, G.; Corradi, G.; Fabbri, F.; Papa, V.; Bandini, E.; Cenacchi, G.; Tazzari, P.L.; et al. An Abnormal Host/Microbiomes Signature of Plasma-Derived Extracellular Vesicles Is Associated to Polycythemia Vera. Front. Oncol. 2021, 11, 715217. [Google Scholar] [CrossRef] [PubMed]
  56. Scherber, R.M.; Langlais, B.T.; Geyer, H.; Dueck, A.; Kosoriek, H.; Johnston, C.; Padrnos, L.; Palmer, J.; Fleischman, A.G.; Mesa, R.A. Nutrition and supplement use characteristics in the myeloproliferative neoplasms: Results from the Nutrient Survey. Blood 2017, 8, 2193. [Google Scholar]
  57. He, Y.; Yue, Y.; Zheng, X.; Zhang, K.; Chen, S.; Du, Z. Curcumin, inflammation, and chronic diseases: How are they linked? Molecules 2015, 20, 9183–9213. [Google Scholar] [CrossRef] [PubMed]
  58. Giordano, A.; Tommonaro, G. Curcumin and Cancer. Nutrients 2019, 11, 2376. [Google Scholar] [CrossRef] [PubMed]
  59. Taverna, S.; Giallombardo, M.; Pucci, M.; Flugy, A.; Manno, M.; Raccosta, S.; Rolfo, C.; De Leo, G.; Alessandro, R. Curcumin inhibits in vitro and in vivo chronic myelogenous leukemia cells growth: A possible role for exosomal disposal of miR-21. Oncotarget 2015, 6, 21918–21933. [Google Scholar] [CrossRef] [PubMed]
  60. Petiti, J.; Rosso, V.; Lo Iacono, M.; Panuzzo, C.; Calabrese, C.; Signorino, E.; Pironi, L.; Cartellà, A.; Bracco, E.; Pergolizzi, B.; et al. Curcumin induces apoptosis in JAK2-mutated cells by the inhibition of JAK2/STAT and mTORC1 pathways. J. Cell. Mol. Med. 2019, 23, 4349–4357. [Google Scholar] [CrossRef] [PubMed]
  61. Hu, Y.; Zhou, N.; Zhu, Q. Curcumin inhibits proliferation and invasion of papillary thyroid carcinoma cells by inhibiting the JAK2/STAT3 pathway. J. BUON 2021, 26, 1635–1641. [Google Scholar] [PubMed]
  62. Duran, C. Available online: https://clinicaltrials.gov/study/NCT06063486 (accessed on 1 May 2025).
  63. Pardanani, A.; Drake, M.T.; Finke, C.; Lasho, T.L.; Rozell, S.A.; Jimma, T.; Tefferi, A. Vitamin D insufficiency in myeloproliferative neoplasms and myelodysplastic syndromes: Clinical correlates and prognostic studies. Am. J. Hematol. 2011, 86, 1013–1016. [Google Scholar] [CrossRef] [PubMed]
  64. Effat, H.; Abohashem, R.S.; Sharaky, M.; Mohammed, M.A. Vitamin D promotes anticancer effects of low-concentration cisplatin-treated non-small cell lung cancer cells via inhibiting the JAK2/STAT3 and TGF-β/SMAD4 pathways. Arch Pharm. 2025, 358, e2400933. [Google Scholar] [CrossRef] [PubMed]
  65. Wakahashi, K.; Minagawa, K.; Kawano, Y.; Kawano, H.; Suzuki, T.; Ishii, S.; Sada, A.; Asada, N.; Sato, M.; Kato, S.; et al. Vitamin D receptor-mediated skewed differentiation of macrophages initiates myelofibrosis and subsequent osteosclerosis. Blood 2019, 133, 1619–1629. [Google Scholar] [CrossRef] [PubMed]
  66. Yetgin, S.; Ozsoylu, S.; Ruacan, S.; Tekinalp, G.; Sarialioğlu, F. Vitamin D-deficiency rickets and myelofibrosis. J. Pediatr. 1989, 114, 213–217. [Google Scholar] [CrossRef] [PubMed]
  67. Irani, M.; Seifer, D.B.; Grazi, R.V.; Julka, N.; Bhatt, D.; Kalgi, B.; Irani, S.; Tal, O.; Lambert-Messerlian, G.; Tal, R. Vitamin D Supplementation Decreases TGF-β1 Bioavailability in PCOS: A Randomized Placebo-Controlled Trial. J. Clin. Endocrinol. Metab. 2015, 100, 4307–4314. [Google Scholar] [CrossRef] [PubMed]
  68. Mohammad, S.; Mishra, A.; Ashraf, M.Z. Emerging Role of Vitamin D and its Associated Molecules in Pathways Related to Pathogenesis of Thrombosis. Biomolecules 2019, 9, 649. [Google Scholar] [CrossRef] [PubMed]
  69. Kucukay, M.B.; Alanli, R. Vitamin D Replacement Effect on Platelet Counts. J. Coll. Physicians Surg. Pak. 2021, 31, 1064–1068. [Google Scholar] [CrossRef] [PubMed]
  70. Ishihara, T.; Yoshida, M.; Arita, M. Omega-3 fatty acid-derived mediators that control inflammation and tissue homeostasis. Int. Immunol. 2019, 31, 559–567. [Google Scholar] [CrossRef] [PubMed]
  71. Dyall, S.C.; Balas, L.; Bazan, N.G.; Brenna, J.T.; Chiang, N.; da Costa Souza, F.; Dalli, J.; Durand, T.; Galano, J.M.; Lein, P.J.; et al. Polyunsaturated fatty acids and fatty acid-derived lipid mediators: Recent advances in the understanding of their biosynthesis, structures, and functions. Prog. Lipid Res. 2022, 86, 101165. [Google Scholar] [CrossRef] [PubMed]
  72. Varney, M.E.; Hardman, W.E.; Sollars, V.E. Omega 3 fatty acids reduce myeloid progenitor cell frequency in the bone marrow of mice and promote progenitor cell differentiation. Lipids Health Dis. 2009, 8, 9. [Google Scholar] [CrossRef] [PubMed]
  73. Javaid, M.; Kadhim, K.; Bawamia, B.; Cartlidge, T.; Farag, M.; Alkhalil, M. Bleeding Risk in Patients Receiving Omega-3 Polyunsaturated Fatty Acids: A Systematic Review and Meta-Analysis of Randomized Clinical Trials. J. Am. Heart Assoc. 2024, 13, e032390. [Google Scholar] [CrossRef] [PubMed]
  74. Marty, C.; Lacout, C.; Droin, N.; Le Couédic, J.P.; Ribrag, V.; Solary, E.; Vainchenker, W.; Villeval, J.L.; Plo, I. A role for reactive oxygen species in JAK2 V617F myeloproliferative neoplasm progression. Leukemia 2013, 27, 2187–2195. [Google Scholar] [CrossRef] [PubMed]
  75. Saddadi, F.; Alatab, S.; Pasha, F.; Ganji, M.R.; Soleimanian, T. The effect of treatment with N-acetylcysteine on the serum levels of C-reactive protein and interleukin-6 in patients on hemodialysis. Saudi J. Kidney Dis. Transpl. 2014, 25, 66–72. [Google Scholar] [CrossRef] [PubMed]
  76. Paterson, R.L.; Galley, H.F.; Webster, N.R. The effect of N-acetylcysteine on nuclear factor-κB activation, interleukin-6, interleukin-8, and intercellular adhesion molecule-1 expression in patients with sepsis. Crit. Care Med. 2003, 31, 2574–2578. [Google Scholar] [CrossRef] [PubMed]
  77. Mokra, D.; Mokry, J.; Barosova, R.; Hanusrichterova, J. Advances in the Use of N-Acetylcysteine in Chronic Respiratory Diseases. Antioxidants 2023, 12, 1713. [Google Scholar] [CrossRef] [PubMed]
  78. Jia, L.; Song, Q.; Zhou, C.; Li, X.; Pi, L.; Ma, X.; Li, H.; Lu, X.; Shen, Y. Dihydroartemisinin as a Putative STAT3 Inhibitor, Suppresses the Growth of Head and Neck Squamous Cell Carcinoma by Targeting Jak2/STAT3 Signaling. PLoS ONE 2016, 11, e0147157. [Google Scholar] [CrossRef] [PubMed]
  79. Li, S.; Xu, W.; Wang, H.; Tang, T.; Ma, J.; Cui, Z.; Shi, H.; Qin, T.; Zhou, H.; Li, L.; et al. Ferroptosis plays an essential role in the antimalarial mechanism of low-dose dihydroartemisinin. Biomed. Pharmacother. 2022, 148, 112742. [Google Scholar] [CrossRef] [PubMed]
  80. Yang, R.; Yan, S.; Zhu, X.; Li, C.Y.; Liu, Z.; Xiong, J.W. Antimalarial drug artemisinin depletes erythrocytes by activating apoptotic pathways in zebrafish. Exp. Hematol. 2015, 43, 331–341.e8. [Google Scholar] [CrossRef] [PubMed]
  81. Dolivo, D.; Weathers, P.; Dominko, T. Artemisinin and artemisinin derivatives as anti-fibrotic therapeutics. Acta Pharm. Sin. B 2021, 11, 322–339. [Google Scholar] [CrossRef] [PubMed]
  82. Premnath, N.; Chung, S.S.; Weinberg, O.K.; Ikpefan, R.; Pandey, M.; Kaur, G.; Geethakumari, P.R.; Afrough, A.; Awan, F.T.; Anderson, L.D., Jr.; et al. Clinical and molecular characteristics associated with Vitamin C deficiency in myeloid malignancies; real world data from a prospective cohort. Leuk. Res. 2023, 125, 107001. [Google Scholar] [CrossRef] [PubMed]
  83. Xie, Z.; Fernandez, J.; Lasho, T.; Finke, C.; Amundson, M.; McCullough, K.B.; LaPlant, B.R.; Mangaonkar, A.A.; Gangat, N.; Reichard, K.K.; et al. High-dose IV ascorbic acid therapy for patients with CCUS with TET2 mutations. Blood 2024, 144, 2456–2461. [Google Scholar] [CrossRef] [PubMed]
  84. Zhao, H.; Zhu, H.; Huang, J.; Zhu, Y.; Hong, M.; Zhu, H.; Zhang, J.; Li, S.; Yang, L.; Lian, Y.; et al. The synergy of Vitamin C with decitabine activates TET2 in leukemic cells and significantly improves overall survival in elderly patients with acute myeloid leukemia. Leuk. Res. 2018, 66, 1–7. [Google Scholar] [CrossRef] [PubMed]
  85. Gillberg, L.; Ørskov, A.D.; Nasif, A.; Ohtani, H.; Madaj, Z.; Hansen, J.W.; Rapin, N.; Mogensen, J.B.; Liu, M.; Dufva, I.H.; et al. Oral vitamin C supplementation to patients with myeloid cancer on azacitidine treatment: Normalization of plasma vitamin C induces epigenetic changes. Clin. Epigenetics 2019, 11, 143. [Google Scholar] [CrossRef] [PubMed]
  86. Tefferi, A.; Pardanani, A.; Lim, K.H.; Abdel-Wahab, O.; Lasho, T.L.; Patel, J.; Gangat, N.; Finke, C.M.; Schwager, S.; Mullally, A.; et al. TET2 mutations and their clinical correlates in polycythemia vera, essential thrombocythemia and myelofibrosis. Leukemia 2009, 23, 905–911. [Google Scholar] [CrossRef] [PubMed]
  87. Mohammed, B.M.; Sanford, K.W.; Fisher, B.J.; Martin, E.J.; Contaifer, D., Jr.; Warncke, U.O.; Wijesinghe, D.S.; Chalfant, C.E.; Brophy, D.F.; Fowler Iii, A.A.; et al. Impact of high dose vitamin C on platelet function. World J. Crit. Care Med. 2017, 6, 37–47. [Google Scholar] [CrossRef] [PubMed]
  88. Pham, T.N.D.; Stempel, S.; Shields, M.A.; Spaulding, C.; Kumar, K.; Bentrem, D.J.; Matsangou, M.; Munshi, H.G. Quercetin Enhances the Anti-Tumor Effects of BET Inhibitors by Suppressing hnRNPA1. Int. J. Mol. Sci. 2019, 20, 4293. [Google Scholar] [CrossRef] [PubMed]
  89. Reyes-Farias, M.; Carrasco-Pozo, C. The Anti-Cancer Effect of Quercetin: Molecular Implications in Cancer Metabolism. Int. J. Mol. Sci. 2019, 20, 3177. [Google Scholar] [CrossRef] [PubMed]
  90. Aghababaei, F.; Hadidi, M. Recent Advances in Potential Health Benefits of Quercetin. Pharmaceuticals 2023, 16, 1020. [Google Scholar] [CrossRef] [PubMed]
  91. Guo, X.; Li, Y.; Wang, W.; Wang, L.; Hu, S.; Xiao, X.; Hu, C.; Dai, Y.; Zhang, Y.; Li, Z.; et al. The construction of preclinical evidence for the treatment of liver fibrosis with quercetin: A systematic review and meta-analysis. Phytother. Res. 2022, 36, 3774–3791. [Google Scholar] [CrossRef] [PubMed]
  92. Nambiar, A.; Kellogg, D., III; Justice, J.; Goros, M.; Gelfond, J.; Pascual, R.; Hashmi, S.; Masternak, M.; Prata, L.; LeBrasseur, N.; et al. Senolytics dasatinib and quercetin in idiopathic pulmonary fibrosis: Results of a phase I, single-blind, single-center, randomized, placebo-controlled pilot trial on feasibility and tolerability. EbioMedicine 2023, 90, 104481. [Google Scholar] [CrossRef] [PubMed]
  93. Tang, Y.; Cheng, S.; Wang, J.; Jin, Y.; Yang, H.; Lin, Q.; Xu, S.; Hui, L.; Yin, Q.; Yang, Y.; et al. Acupuncture for the Treatment of Itch: Peripheral and Central Mechanisms. Front Neurosci. 2022, 15, 786892. [Google Scholar] [CrossRef] [PubMed]
  94. Kim, E.; Hur, S.; Hwang, C.; Jang, E.; Lee, J.; Kim, Y. A Case Report on Symptom Improvement in a Polycythemia Vera Patient Treated with Acupuncture. J. Int. Korean Med. 2021, 42, 976–981. [Google Scholar] [CrossRef]
  95. Kumada, K.; Matsumoto-Miyazaki, J.; Okada, H.; Okura, H.; Sato, Y. Successful Administration of Kampo Medicine and Acupuncture Treatment to Improve Erythromelalgia: A Case Report. Cureus 2024, 16, 7–12. [Google Scholar] [CrossRef] [PubMed]
  96. 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]
  97. Levy Yurkovski, I.; Andreazzoli, F.; Ben-Arye, E.; Attias, S.; Tadmor, T. Integrative Approaches in the Treatment of Patients Affected by Lymphoma. Curr. Oncol. Rep. 2023, 25, 1523–1534. [Google Scholar] [CrossRef] [PubMed]
  98. El Iskandarani, S.; Deng, G. Acupuncture in hematologic malignancies and hematopoietic cell transplantation. Blood Rev. 2022, 56, 100985. [Google Scholar] [CrossRef] [PubMed]
  99. Frenkel, M.; Sapire, K. Complementary and Integrative Medicine in Hematologic Malignancies: Questions and Challenges. Curr. Oncol. Rep. 2017, 19, 79. [Google Scholar] [CrossRef] [PubMed]
  100. Acupuncture Association of Chartered Physiotherapists (AACP). AACP Guidelines for Safe Practice of Acupuncture. 2017. Available online: https://www.aacp.org.uk/assets/ckfinder_library/files/AACP%20Safety%20Guidelines%202017%20online.pdf (accessed on 27 March 2025).
  101. Health Centre. Infection Risk with Acupuncture. Available online: https://www.healthcentre.org.uk/acupuncture/infection-risk-with-acupuncture.html (accessed on 27 March 2025).
  102. Kayo, T.; Suzuki, M.; Mitsuma, T.; Suzuki, M.; Ikeda, S.; Sukegawa, M.; Tsunoda, S.; Ohta, M. Bleeding Risk of Acupuncture for Patients with Hematological Malignancies Accompanying Thrombocytopenia: A Retrospective Chart Review. J. Integr. Complement. Med. 2024, 30, 77–84. [Google Scholar] [CrossRef] [PubMed]
  103. Eckert, R.; Huberty, J.; Gowin, K.; Mesa, R.; Marks, L. Physical activity as a nonpharmacological symptom management approach in myeloproliferative neoplasms: Recommendations for future research. Integr. Cancer Ther. 2017, 16, 439–450. [Google Scholar] [CrossRef] [PubMed]
  104. Cormie, P.; Zopf, E.M.; Zhang, X.; Schmitz, K.H. The impact of exercise on cancer mortality, recurrence, and treatment-related adverse effects. Epidemiol. Rev. 2017, 39, 71–92. [Google Scholar] [CrossRef] [PubMed]
  105. 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]
  106. Ballard-Barbash, R.; Friedenreich, C.M.; Courneya, K.S.; Siddiqi, S.M.; McTiernan, A.; Alfano, C.M. Physical activity, biomarkers, and disease outcomes in cancer survivors: A systematic review. J. Natl. Cancer Inst. 2012, 104, 815–840. [Google Scholar] [CrossRef] [PubMed]
  107. Shephard, R.J.; Shek, P.N. Effects of exercise and training on natural killer cell counts and cytolytic activity: A meta-analysis. Sports Med. 1999, 28, 177–195. [Google Scholar] [CrossRef] [PubMed]
  108. Idorn, M.; thor Straten, P. Exercise and cancer: From “healthy” to “therapeutic”? Cancer Immunol. Immunother. 2017, 66, 667–671. [Google Scholar] [CrossRef] [PubMed]
  109. Pedersen, L.; Christensen, J.F.; Hojman, P. Effects of exercise on tumor physiology and metabolism. Cancer J. 2015, 21, 111–116. [Google Scholar] [CrossRef] [PubMed]
  110. Löf, M.; Bergström, K.; Weiderpass, E. Physical activity and biomarkers in breast cancer survivors: A systematic review. Maturitas 2012, 73, 134–142. [Google Scholar] [CrossRef] [PubMed]
  111. Courneya, K.S.; Segal, R.J.; Mackey, J.R.; Gelmon, K.; Reid, R.D.; Friedenreich, C.M.; Ladha, A.B.; Proulx, C.; Vallance, J.K.; Lane, K.; et al. Effects of aerobic and resistance exercise in breast cancer patients receiving adjuvant chemotherapy: A multicenter randomized controlled trial. J. Clin. Oncol. 2007, 25, 4396–4404. [Google Scholar] [CrossRef] [PubMed]
  112. Denham, J.; Marques, F.Z.; O’Brien, B.J.; Charchar, F.J. Exercise: Putting action into our epigenome. Sports Med. 2014, 44, 189–209. [Google Scholar] [CrossRef] [PubMed]
  113. Chen, Y.W.; Apostolakis, S.; Lip, G.Y. Exercise-induced changes in inflammatory processes: Implications for thrombogenesis in cardiovascular disease. Ann. Med. 2014, 46, 439–455. [Google Scholar] [CrossRef] [PubMed]
  114. Tolstrup Larsen, R.; Tang, L.H.; Brochmann, N.; Meulengracht Flachs, E.; Illemann Christensen, A.; Hasselbalch, H.C.; Zwisler, A.D. Associations between fatigue, physical activity, and QoL in patients with myeloproliferative neoplasms. Eur. J. Haematol. 2018, 100, 550–559. [Google Scholar] [CrossRef] [PubMed]
  115. Scherber, R.M.; Kosiorek, H.E.; Senyak, Z.; Dueck, A.C.; Clark, M.M.; Boxer, M.A.; Geyer, H.L.; McCallister, A.; Cotter, M.; Van Husen, B.; et al. Comprehensively understanding fatigue in patients with myeloproliferative neoplasms. Cancer 2016, 122, 477–485. [Google Scholar] [CrossRef] [PubMed]
  116. Gowin, K.L.; Langlais, B.T.; Millstine, D.; Kosiorek, H.E.; Huberty, J.; Eckert, R.; Mesa, R.A. Survey of Integrative Medicine in Myeloproliferative Neoplasms (The SIMM Study-2). Blood 2018, 132, 3047. [Google Scholar] [CrossRef]
  117. Pedersen, K.M.; Zangger, G.; Brochmann, N.; Grønfeldt, B.M.; Zwisler, A.D.; Hasselbalch, H.C.; Tang, L.H. The effectiveness of exercise-based rehabilitation to patients with myeloproliferative neoplasms—An explorative study. Eur. J. Cancer Care 2018, 27, e12865. [Google Scholar] [CrossRef] [PubMed]
  118. Ligibel, J.A.; Denlinger, C.S. New NCCN guidelines® for survivorship care. J. Natl. Compr. Cancer Netw. 2013, 11, 640–644. [Google Scholar] [CrossRef] [PubMed]
  119. Felser, S.; Rogahn, J.; Hollenbach, L.; Gruen, J.; le Coutre, P.; Al-Ali, H.K.; Schulze, S.; Muegge, L.O.; Kraze-Kliebhahn, V.; Junghanss, C. Physical exercise recommendations for patients with polycythemia vera based on preferences identified in a large international patient survey study of the East German Study Group for Hematology and Oncology (OSHO# 97). Cancer Med. 2023, 12, 18235–18245. [Google Scholar] [CrossRef] [PubMed]
  120. Felser, S.; Rogahn, J.; le Coutre, P.; Al-Ali, H.K.; Schulze, S.; Muegge, L.O.; Gruen, J.; Geissler, J.; Kraze-Kliebhahn, V.; Junghanss, C. Anxieties, age and motivation influence physical activity in patients with myeloproliferative neoplasms-a multicenter survey from the East German study group for hematology and oncology (OSHO# 97). Front. Oncol. 2023, 12, 1056786. [Google Scholar]
  121. Mind-Body Practice. Available online: https://www.cancer.gov/publications/dictionaries/cancer-terms/def/mind-body-practice (accessed on 1 May 2025).
  122. Deleemans, J.M.; Mather, H.; Spiropoulos, A.; Toivonen, K.; Baydoun, M.; Carlson, L.E. Recent progress in mind–body therapies in Cancer Care. Curr. Oncol. Rep. 2023, 25, 293–307. [Google Scholar] [CrossRef] [PubMed]
  123. Bower, J.E.; Irwin, M.R. Mind–body therapies and control of inflammatory biology: A descriptive review. Brain Behav. Immun. 2016, 51, 1–11. [Google Scholar] [CrossRef] [PubMed]
  124. Carney, L.M.; Park, C.L.; Hingorany, P. The mechanisms of mindfulness-based stress reduction and mindfulness-based cognitive therapy for cancer patients and survivors: A systematic review. Psychol. Conscious. Theory Res. Pract. 2023. [Google Scholar] [CrossRef]
  125. Buffart, L.M.; van Uffelen, J.G.; Riphagen, I.I.; Brug, J.; van Mechelen, W.; Brown, W.J.; Chinapaw, M.J. Physical and psychosocial benefits of yoga in cancer patients and survivors, a systematic review and meta-analysis of randomized controlled trials. BMC Cancer 2012, 12, 559. [Google Scholar] [CrossRef] [PubMed]
  126. Huberty, J.; Eckert, R.; Dueck, A.; Kosiorek, H.; Larkey, L.; Gowin, K.; Mesa, R. Online yoga in myeloproliferative neoplasm patients: Results of a randomized pilot trial to inform future research. BMC Complement. Altern. Med. 2019, 19, 121. [Google Scholar] [CrossRef] [PubMed]
  127. Huberty, J.; Eckert, R.; Larkey, L.; Gowin, K.; Mitchell, J.; Mesa, R. Perceptions of myeloproliferative neoplasm patients participating in an online yoga intervention: A qualitative study. Integr. Cancer Ther. 2018, 17, 1150–1162. [Google Scholar] [CrossRef] [PubMed]
  128. Huberty, J.; Eckert, R.; Gowin, K.; Mitchell, J.; Dueck, A.C.; Ginos, B.F.; Larkey, L.; Mesa, R. Feasibility study of online yoga for symptom management in patients with myeloproliferative neoplasms. Haematologica 2017, 102, e384. [Google Scholar] [CrossRef] [PubMed]
  129. Eckert, R.; Huberty, J.; Dueck, A.; Kosiorek, H.; Larkey, L.; Mesa, R.A. A pilot study of online yoga to improve fatigue and quality of life in myeloproliferative neoplasm patients. Blood 2017, 130, 3443. [Google Scholar]
  130. Eckert, R.; Huberty, J.; Gowin, K.L.; Ginos, B.; Kosiorek, H.E.; Dueck, A.C.; Mesa, R.A. Impact of weight on symptom burden outcomes in myeloproliferative neoplasm patients participating in an online yoga intervention. Blood 2016, 128, 5481. [Google Scholar] [CrossRef]
  131. van Waart, H.; van Harten, W.H.; Buffart, L.M.; Sonke, G.S.; Stuiver, M.M.; Aaronson, N.K. Why do patients choose (not) to participate in an exercise trial during adjuvant chemotherapy for breast cancer? Psycho-Oncology 2016, 25, 964–970. [Google Scholar] [CrossRef] [PubMed]
  132. Kabat-Zinn, J. Living FC. Using the Wisdom of Your Body and Mind to Face Stress, Pain, and Illness; Bantam Doubleday Dell: New York, NY, USA, 1990; p. 2383411. [Google Scholar]
  133. Huberty, J.; Eckert, R.; Larkey, L.; Kurka, J.; De Jesús, S.A.; Yoo, W.; Mesa, R. Smartphone-based meditation for myeloproliferative neoplasm patients: Feasibility study to inform future trials. JMIR Form. Res. 2019, 3, e12662. [Google Scholar] [CrossRef] [PubMed]
  134. Win, H.; Russell, S.; Wertheim, B.C.; Maizes, V.; Crocker, R.; Brooks, A.J.; Mesa, R.; Huberty, J.; Geyer, H.; Eckert, R.; et al. Mobile app intervention on reducing the myeloproliferative neoplasm symptom burden: Pilot feasibility and acceptability study. JMIR Form. Res. 2022, 6, e33581. [Google Scholar] [CrossRef] [PubMed]
  135. Puzia, M.E.; Huberty, J.; Eckert, R.; Larkey, L.; Mesa, R. Associations between global mental health and response to an app-based meditation intervention in myeloproliferative neoplasm patients. Integr. Cancer Ther. 2020, 19, 1534735420927780. [Google Scholar] [CrossRef] [PubMed]
  136. Huberty, J.; Eckert, R.; Larkey, L.; Joeman, L.; Mesa, R. Experiences of using a consumer-based mobile meditation app to improve fatigue in myeloproliferative patients: Qualitative study. JMIR Cancer 2019, 5, e14292. [Google Scholar] [CrossRef] [PubMed]
  137. Fincham, G.W.; Mavor, K.; Dritschel, B. Effects of mindfulness meditation duration and type on well-being: An online dose-ranging randomized controlled trial. Mindfulness 2023, 14, 1171–1182. [Google Scholar] [CrossRef] [PubMed]
Figure 1. A potential integrative approach in MPNs.
Figure 1. A potential integrative approach in MPNs.
Jcm 14 05080 g001
Table 1. Potential integrative approaches and research directions in MPN management in myeloproliferative neoplasms (MPNs).
Table 1. Potential integrative approaches and research directions in MPN management in myeloproliferative neoplasms (MPNs).
ModalityMechanismContraindicationsResearch Priorities
Disease Management
Mediterranean dietAnti-inflammatory, ↓CV risk [23,24]SafeRCT: inflammation markers, BMI, QOL [5,39]
Vitamin D↓Thrombosis risk [68]; JAK/STAT interference [64]BM fibrosis interaction [65,66,67]Dosing studies, serum monitoring
CurcuminCurcumin JAK/STAT inhibitor [57,58,59,60,61]Anticoagulant interactionSafety/efficacy trials, drug interactions
Omega-3Anti-inflammatory [71]; ↓hypercoagulability [71]SafeOptimal dosing, bleeding risk assessment
NACAntioxidant [74]; ↓ROS, NFκB [75,76]SafeIV vs. oral trials, dose-response
ArtemisininJAK/STAT inhibitor [78,79]Low toxicityPhase I/II trials in MPN
QuercetinJAK2/BET modulator [84,85,86]UnknownSafety/efficacy trials, drug interactions
Vitamin CUnclear in MPNMay worsen PV [87]Safety/efficacy trials
Symptom Management
Mindfulness↑QOL, symptom relief [133,134]Technical limitations [135,137]RCT: standardized protocols, QOL
Yoga↓TNF [130]Spleen irritation [131]Protocols for MPN
Physical Activity↓Fatigue, ↑strength [116,117]SafeExercise prescription trials
AcupuncturePruritus, fatigue, pain relief [93,94,95,96,97,98,99]Bleeding/infection risk [100,101]RCT: inflammation markers, QOL
Touch/Music/ArtSymptom relief, mood, QOLSafeComparative effectiveness research
Abbreviations: BET = bromodomain and extra-terminal motif; CV = cardiovascular; JAK/STAT = Janus kinase/signal transducer and activator of transcription; NAC = N-acetylcysteine; PV = polycythemia vera; QOL = quality of life; ROS = reactive oxygen species; ↓ = reduced; ↑ = increased.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Andreazzoli, F.; Levy Yurkovski, I.; Gowin, K.; Bonucci, M. Management of Myeloproliferative Neoplasms: An Integrative Approach. J. Clin. Med. 2025, 14, 5080. https://doi.org/10.3390/jcm14145080

AMA Style

Andreazzoli F, Levy Yurkovski I, Gowin K, Bonucci M. Management of Myeloproliferative Neoplasms: An Integrative Approach. Journal of Clinical Medicine. 2025; 14(14):5080. https://doi.org/10.3390/jcm14145080

Chicago/Turabian Style

Andreazzoli, Francesca, Ilana Levy Yurkovski, Krisstina Gowin, and Massimo Bonucci. 2025. "Management of Myeloproliferative Neoplasms: An Integrative Approach" Journal of Clinical Medicine 14, no. 14: 5080. https://doi.org/10.3390/jcm14145080

APA Style

Andreazzoli, F., Levy Yurkovski, I., Gowin, K., & Bonucci, M. (2025). Management of Myeloproliferative Neoplasms: An Integrative Approach. Journal of Clinical Medicine, 14(14), 5080. https://doi.org/10.3390/jcm14145080

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