Introduction
The subtle imbalance of inflammatory and anti-inflammatory signals observed in noncommunicable diseases (NCDs), especially as people age, is called “metainflammation” (or “inflammaging”).1 Metainflammation involves increased inflammatory markers such as tumor necrosis factor-α, interferon-β, interleukin (IL)-1, IL-6, IL-8, IL-12, IL-17, IL-22, chemokines, C-reactive protein, and monocyte chemoattractant protein-1.2 These inflammatory signals contribute to the development of NCDs, a name given to chronic, non transmisibles diseases like cancer,3,4 cardiovascular disorders,5 metabolic syndrome,6,7 diabetes,8,9 neurodegenerative disorders,10,11 and renal12 and lung13 diseases. Globally NCDs are responsible for 74% of all deaths,14 with 17 million occurring before 70 years of age; 86% of these deaths occur in low developed countries.
NCDs involve alterations in pro- and anti-inflammatory pathways. Major proinflammatory pathways include nuclear factor-κB15 and cyclooxygenase,16 while anti-inflammatory pathways involve specialized pro-resolving mediators17,18 and the melatonin / endocannabinoid / angiotensin 1,7 axis pathways.19,20 The body’s ability to cope with inflammation decreases with age, thus favoring proinflammatory processes.1 As the global population ages, health challenges increase, including the prevalence of NCDs.21–23
Despite numerous studies in the psycho-neuro-endocrine-immunology field demonstrating that the nervous, immune and endocrine systems are not physiologically segregated and cannot be considered as isolated entities,24–26 the diagnosis and therapies of NCDs continue to be separately carried out in the clinical practice.27 This separation between experimental and clinical medicine has become questioned by the discovery of human endogenous molecules providing anti-inflammatory and antitumor activity in NCDs. Showing very few or no toxicity, three fundamental classes constitute the core of a possible therapeutic approach to treat NCDs, ie, the endogenous anti-inflammatory and antitumor network given by pineal methoxyindoles,28 endocannabinoids,29 and angiotensin 1–7.30
We hereby report a retrospective cross-sectional study of 306 adult patients with NCDs to assess the therapeutic efficacy and safety of a neuroimmune regimen containing the above mentioned antiinflammatory compounds. This neuroimmune regimen (melatonin, 5-methoxytryptamine, endocannabinoids, and angiotensin 1–7) was tested in patients with advanced, untreatable cancers, autoimmune diseases, neurodegenerative pathologies, neuropsychiatric disorders, and cardiopulmonary pathologies.
Patients and Methods
From March 2020 to April 2023, 306 patients with systemic diseases were evaluated (M/F: 141/165; median age: 64 years; range: 16–92 years). This is a single arm interventional study (before-and-after study) to evaluate the effectiveness of the neuroimmune regimen by comparing outcomes measured before and after the intervention is applied.
Eligibility criteria for cancer patients were: 1) histologically proven neoplasms and measurable lesions assessed by The WHO clinical classification of tumors31 and by performing CT scan, NMR, or PET every 3-months; 2) lack of response to the standard medical oncological therapies, including the most widely used chemotherapy, endocrine treatment, anti-angiogenic therapy, targeted and immunotherapies, or progression under treatment; 3) a life expectancy less than 1 year (mostly less than 6 months); 4) no other concomitant complementary cancer therapies.
As far as patients with other systemic diseases, the eligible criteria were the existence of clinically evaluable parameters, consisting of autoantibody titer for patients with autoimmune disease, cognitive tests and neuropsychiatric clinical investigation for neuropsychiatric patients, and the common clinical evaluations of the cardio-pulmonary functions in patients with cardio-pulmonary pathologies.
The exclusion criteria were chronic therapy with pregabalin because of its inhibitory action on angiotensin 1–7 activity32 or with fentanyl or other major opiates, because of their important immunosuppressive action on anticancer immunity.33
The WHO clinical classification of tumors comprises several response categories to evaluate the effectiveness of treatments in oncology.31 They include: Complete Response (CR) indicating the disappearance of all signs of cancer. Partial Response (PR) indicating a significant reduction in tumor size or extent of metastasis, but not a complete disappearance. Typically, this means a decrease of at least 30% in the size of the measurable tumor lesions. Stable Disease (SD): This classification is used when the cancer has neither decreased significantly in size nor increased. Essentially, the disease remains unchanged, indicating that the treatment has halted the progression of the disease but has not reduced the tumor size. Disease Control (DC): This term encompasses both complete and partial responses, as well as stable disease. It indicates that the treatment has been effective in controlling the disease, preventing further progression. Progressive Disease (PD): This indicates that the cancer has increased in size or spread to new areas despite treatment.
After the approval of the local Ethical Committee of the Institute of Biological Medicine (acta no. 01/10, dated February 1, 2020), the clinical protocol was explained to each patient, particularly emphasizing the novelty of using natural molecules drawn from human body itself, and a written consent was obtained in each case. All procedures were conducted in accordance with the Declaration of Helsinki.
The neuroimmune regimen included the oral administration of angiotensin 1–7 0.5 mg twice/day (8.00 A.M. and 8.00 P.M) in association with two major oncostatic pineal hormones (melatonin 100 mg/day at bedtime, 5-methoxytryptamine 30 mg/day in the early afternoon), and with cannabidiol at a dose of 20 mg twice/day. For patients affected by primary or relapsed glioblastoma, a third pineal oncostatic hormone was administered (beta-carboline 6-methoxy-1,2,3,4-tetra-hydro-beta-carboline, pinoline) at a dose of 1 mg at 8.00 P.M.34 In the group of hypertensive patients, six of them were already under treatment with anti-hypertensive agents, while the remaining eight patients started with the neuroimmune regimen for personal decision. In patients suffering from depression, the protocol included the oral administration of oxytocin at a dose of 1–2 mg twice/day because of its documented efficacy in improving mood and in the relief of anxiety.35 The neuroimmune regimen was continued without interruption until disease progression. Patients were also investigated for their changes in the immunobiological response in relation to the clinical course of their pathology, and the immune status was assessed by determining the values of lymphocyte-to-monocyte ratio (LMR), whose progressive decline predicts a poor prognosis either in cancer36 or in cardiovascular and pulmonary diseases.37 LMR has been shown to be a significant prognostic marker in several types of cancer, including glioma, breast cancer, and small cell lung cancer. LMR reflects the balance between lymphocytes, which are crucial for adaptive immune responses, and monocytes, which are involved in inflammation and tissue repair. A low LMR indicates a higher inflammatory state and a potentially compromised immune response. Normal values of LMR observed in our laboratory were higher than 2.1 (95% confidence limits) and were established on the basis of the values found in a control group of 300 age-and sex-matched healthy subjects. LMR was evaluated before the onset of treatment, and at monthly intervals.
Data were reported as frequency (%) or mean ± SEM, as appropriate. After verifying for normality for distribution, paired t-test were used for LMR evaluation. In addition, as far as oncological patients, the 1-year percent of survival was evaluated using the Kaplan-Meier method and Log rank test in a group of 188 advanced cancer patients who received the neuroimmune regimen as compared to a historical control group of 200 age-, sex-, and histotype-matched untreatable cancer patients, who were treated with only palliative therapy.
Results
Table 1 summarizes the results of the neuroimmune regimen used in 306 patients with noncommunicable diseases (NCDs) refractory to standard therapies. Among patients with advanced, untreatable solid tumors, a complete response (CR) was observed in 8/188 (4%) of treated cancer patients. A partial response (PR) was achieved in 31/188 (12%) cancer patients. Thus, tumor regression was found in 39/188 (16%) patients. A stable disease (SD) was observed in 97/188 (52%) patients. Therefore, a disease control (DC) (CR + PR + SD) was verified in 128/188 (68%) patients as far as all solid tumor types, whereas the remaining 60/188 (32%) patients showed a progressive disease (PD). Concerning hematologic tumors (N= 11), values of CR, PR, SD, DC and PD were 0, 1 (9%), 1 (9%), 6 (55%), 7 (64%) and 4 (36%) (Table 1).
Table 1 Melatonin + 5-Methoxytryptamine + Angiotensin 1–7 + Cannabidiol Treatment in 306 Patients With Noncommunicable Diseases (NCDs) Refractory to Standard Therapies
The mean duration of the oncostatic response was 14 months (range 3–36+ months). A remarkable result was observed in patients with glioblastoma, with an objective tumor regression in 10/48 (21%) patients, and a DC in 35/48 (73%) patients. Notable results were also obtained in other malignant human neoplasms, like pancreatic adenocarcinoma [DC: 10/19 (53%) patients] and triple negative breast cancer [DC: 13/19 (68%) patients]. A survival longer than 1 year with respect to a life expectancy less than one year (less than 6 months in most cases) was achieved in 135/188 (72%) cancer patients.
The 1-year survival curves observed in the treated group of cancer patients and in the control group are illustrated in Figure 1. The percent of 1-year survival obtained in patients treated by the neuroendocrine regimen was significantly higher than in the control group treated by the only best supportive care (P<0.005).
Figure 1 Percent of 1-year survival observed in 188 advanced cancer patients who received the neuroimmune regimen including pineal methoxyindoles, angiotensin 1–7, and endocannabinoids, as compared to a historical control group of 200 age-, sex-, and histotype-matched untreatable cancer patients, who were treated with only palliative therapy. Differences between groups are significant (P< 0.005), Kaplan-Meier method and Log rank test.
Clinical benefits were also obtained in patients suffering from NCDs other than cancer. A particular efficacy of the neuroimmune regimen was observed in the treatment of autoimmune diseases, including multiple sclerosis, Hashimoto’s thyroiditis, and other autoimmune pathologies. The control of neurological symptoms was verified in 13/17 (76%) patients with multiple sclerosis, a decline in anti-thyroglobulin antibody levels in 6/7 (86%) Hashimoto’s thyroiditis cases, and a decline in antinuclear antibody production in 9/12 (75%) patients with other autoimmune diseases.
Beneficial results were obtained in 12/19 (63%) patients with neurodegenerative pathologies, including Parkinson’s and Alzheimer’s diseases, as well as in one patient affected by the motoneuron pathology. Depressive patients were particularly responsive to oxytocin treatment in association with the neuroimmune regimen, with a clear improvement in mood in 7/10 (70%) patients.
A reversion in insulin resistance, as shown by a progressive decline in hemoglobin glycate blood concentrations, was verified in 9/12 (75%) diabetic patients. In addition, patients affected by cardiovascular or pulmonary pathologies were particularly responsive to treatment eg, in idiopathic hypertension a control of blood pressure was achieved in 10/14 (71%) patients within the first two weeks of therapy. A reduction in brain natriuretic peptide (BNP) levels was observed in 6/9 (67%) patients with heart failure.
The treatment was well tolerated in all patients, and no biological toxicity occurred. In more detail, no negative effect was seen on hemoglobin levels, platelet count, cardiac function, and renal and hepatic metabolic activities. Most patients referred an improvement in their life due to a better quality of sleep, mood increase, and relief of anxiety and pain, with a particular efficacy in the treatment of cancer-related asthenia. In addition, all patients showed an increased diuresis, possibly due to the diuretic action of angiotensin 1–7.30
LMR mean values significantly increased on therapy with respect to the pretreatment in patients who showed a clinical benefit (3.6 ± 0.3 vs 2.3 ± 0.2, P<0.05). On the contrary, no meaningful change occurred in patients who had no improvement on treatment (2.8 ± 0.4 vs 2.4 ± 0.3). This difference was particular evident in patients with advanced untreatable cancer as compared to those with refractory, progressive disease (4.3 ± 0.4 vs 2.3 ± 0.2, P<0.01), while no difference was seen as far as their pretreatment values (2.7 ± 0.3 vs 2.3 ± 0.4).
Discussion
This study demonstrates that a neuroimmune regimen incorporating melatonin (100 mg/day at night), 5-methoxytryptamine (30 mg in the early afternoon), angiotensin 1–7 (0.5 mg twice daily), and cannabidiol (20 mg twice daily) may achieve meaningful clinical benefits in patients with severe, treatment-resistant NCDs. These findings underscore the potential of natural, endogenous molecules like pineal hormones, angiotensin 1–7, and endocannabinoids to modulate the immune system, controlling inflammation, cell proliferation, and immune response. Enhancing the immune balance through this regimen, with adjuncts such as a low-dose of IL-2, could be particularly beneficial in NCDs patients.38
Melatonin is a naturally occurring substance found in all living organisms, from single-celled entities to humans. It is produced not only in the pineal gland but also in various tissues throughout the body, where it may serve a protective role through paracrine or autocrine effects.39 Via its anti-inflammatory effects, melatonin supresses tumor growth.40,41 In addition, melatonin efficacy has been demonstrated in several NCDs.42–44 In the case of cancer, melatonin’s antiproliferative mechanisms include, in addition to direct and indirect antioxidant effects, the modulation of cell cycle, anti-angiogenic and antimetastatic effects, induction of apoptosis in cancer cells, a decrease in telomerase activity and effective immune modulation.45 In addition to its cancer-fighting properties, melatonin should be considered for treating non-communicable diseases (NCDs) for two key reasons. Firstly, its hypnotic and chronobiotic properties enable an effective management of sleep disturbances, which are a significant co-morbidity in cancer patients.46 Secondly, because the antidepressant and anxiolytic activity of melatonin,47,48 it is useful to treat depression and anxiety seen in cancer as well as in other NCDs.
In addition to melatonin, the pineal produces other relevant antitumor indole hormones, like 5-methoxytryptamine,49 and beta-carbolines.50,51 This explains why the increase in tumour growth that follows pinealectomy cannot be totally counteracted by the exogenous administration of melatonin. 5-Methoxytryptamine may play a strong oncostatic activity.49 Concerning pineal beta-carbolines, the best known is pinoline,50,51 that together to anti-depressant and anxiolytic effects, it also has anti-inflammatory and antitumor properties.34 A pineal endocrine deficiency in cancer was suggested by histological studies.52
Building on melatonin’s role, another key component of the neuroimmune regimen is angiotensin 1–7, an enzymatic product of angiotensin converting enzyme 2 (ACE2) and regulates immune-inflammatory responses, cell proliferation, blood coagulation, fibrosis, cardiopulmonary functions, and metabolic activities.53–55 Angiotensin 1–7 exhibits effects that contrast sharply with those of angiotensin II, an ACE product known for its inflammatory, tumor-promoting, immunosuppressive, hypertensive, cardiotoxic, neurotoxic, pro-thrombotic, pro-fibrotic, and pro-diabetic activities,56–60 Conversely, angiotensin 1–7 induces hypotensive, diuretic, cardio-neuroprotective, anti-inflammatory, anti-tumoral, anti-thrombotic, anti-fibrotic, and anti-diabetic effects with minimal toxicity.61–64
Positive feedback takes place between angiotensin II and IL-17 released from TH17 lymphocytes.57 On the contrary, angiotensin 1–7 counteracts IL-17 secretion and promotes IL-2 production, a key oncostatic cytokine.30 The antagonistic relationship between angiotensin II and angiotensin 1–7 has implications for cardiovascular health and immune system function, especially considering that angiotensin II can foster cancer onset and metastasis. Indeed, ACE inhibitors or angiotensin II receptor blockers have been linked to a reduced incidence of cancer in hypertensive patients.65,66
The function of the renin - ACE - angiotensin II - ACE2 - angiotensin 1–7 axis depends on the modulation of ACE and ACE2 expression, which is under a central regulation played by the neuroendocrine system. ACE2 is stimulated by melatonin67 and estradiol.68 The progressive age-related decline in both melatonin and estradiol secretion would explain the higher blood levels of angiotensin 1–7 in fertile women than in men, and its lower concentration in aged subjects. In addition to the pineal gland and the ACE2- angiotensin 1–7 axis, a third anti-inflammatory / oncostatic component is given by the endocannabinoid system,69 which is connected to the pineal gland by reciprocal stimulatory actions.70,71
The discovery of ACE2 - angiotensin 1-7 - Mas receptor axis, whose biological effects are opposite to those played by ACE - angiotensin II - AT1R axis, leads to a reinterpretation of the pathogenesis of the cytokine network participating in several biological systems. In the brain, an enhanced expression of ACE leading to enhanced local production of angiotensin II and a decrease of angiotensin 1–7 could represent the first event involved in neuroinflammation.72,73 Depression would be also promoted by an increased brain production of IL-17 and other inflammatory cytokines because of a reduced ACE2 expression at brain sites.74 Lately, ACE / ACE2 ratio would influence the clinical history of the metabolic syndrome, since insulin resistance follows angiotensin II administration and is prevented by angiotensin 1–7 administration.75,76 Despite the evidence suggesting the therapeutic potential of angiotensin 1–7 and melatonin in most human systemic diseases, very few clinical studies have evaluated their therapeutic significance in NCDs.
The present study had several limitations. Retrospective studies are prone to selection bias, recall bias, and confounding factors, which can affect the validity of the results. In a cross-sectional, retrospective analysis, relevant data could be lost, and it does not allow modifications to be made/added to the variables already established. Double-blind, placebo-controlled trials will contribute to further support the hypothesis put forth by minimizing of bias and establishing causality between the intervention and outcome. The use of placebo and control groups will allow for a clear comparison of the effectiveness of the neuroimmune regimen proposed.
Conclusion
This study highlights the potential of leveraging endogenous molecules to treat NCDs by modulating cell proliferation, inflammation, immune responses, metabolism, and neurological functions. The findings suggest that a neuroimmune regimen incorporating melatonin, angiotensin 1–7, and other bioactive compounds could offer a low-cost, minimally toxic therapeutic approach. Longer follow-up and randomized studies by comparing the results with those obtained in patients treated only with palliative therapy are necessary to better establish the impact of the neuroendocrine therapy on the survival of untreatable cancer patients.
Acknowledgments
The authors would like to thank the patients and research assistants for their participation and support.
Author Contributions
All authors made a significant contribution to the work reported, whether in the conception, study design, execution, acquisition of data, analysis and interpretation, or in all these areas; took part in drafting, revising or critically reviewing the article; gave final approval of the version to be published; agreed on the journal to which the article has been submitted; and agreed to be accountable for all aspects of the work.
Funding
The authors declare that no financial support was received for the research, authorship, and/or publication of this article. The study was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
Disclosure
The authors report no conflicts of interest in this work.
1. Fulop T, Larbi A, Pawelec G, et al. Immunology of aging: the birth of inflammaging. Clin Rev Allergy Immunol. 2021;64:109–122. doi:10.1007/S12016-021-08899-6
2. Xia S, Zhang X, Zheng S, et al. An update on inflamm-aging: mechanisms, prevention, and treatment. J Immunol Res. 2016;2016:1–12. doi:10.1155/2016/8426874
3. Munn LL. Cancer and inflammation. Wiley Interdiscip Rev Syst Biol Med. 2017;9(2). doi:10.1002/WSBM.1370
4. Lissoni P. Therapy implications of the role of interleukin-2 in cancer. Expert Rev Clin Immunol. 2017;13(5):491–498. doi:10.1080/1744666X.2017.1245146
5. Zhang Y, Xie Y, Zhang C, et al. The systemic immune inflammation index as a novel predictive biomarker for contrast-induced acute kidney injury risk following percutaneous coronary intervention: a meta-analysis of cohort studies. Curr Vasc Pharmacol. 2024;23. doi:10.2174/0115701611328810241028112700
6. Yi Q, Li X, He Y, et al. Associations of dietary inflammatory index with metabolic syndrome and its components: a systematic review and meta-analysis. Public Health Nutr. 2021;24:5463–5470. doi:10.1017/S1368980021000288
7. Bakhshimoghaddam F, Chaharlang R, Mansoori A, et al. Dietary inflammatory index and its association with risk of metabolic syndrome and its components: a systematic review and meta-analysis of observational studies. J Health Popul Nutr. 2024;43(1). doi:10.1186/S41043-024-00580-W
8. Mohammadpour Fard R, Rashno M, Bahreiny SS. Effects of melatonin supplementation on markers of inflammation and oxidative stress in patients with diabetes: a systematic review and meta-analysis of randomized controlled trials. Clin Nutr ESPEN. 2024;63:530–539. doi:10.1016/j.clnesp.2024.07.015
9. Muzammil K, Khaleel AQ, Merza MS, et al. The effects of ω-3 fatty acids on inflammatory and oxidative stress markers in patients with Type 2 diabetes mellitus: a systematic review and meta-analysis of controlled trials. Prostaglandins Other Lipid Mediat. 2024;175:106887. doi:10.1016/J.PROSTAGLANDINS.2024.106887
10. Chen X, Hu Y, Cao Z, et al. Cerebrospinal fluid inflammatory cytokine aberrations in Alzheimer’s disease, Parkinson’s disease and amyotrophic lateral sclerosis: a systematic review and meta-analysis. Front Immunol. 2018;9. doi:10.3389/fimmu.2018.02122
11. Gautam AS, Pulivarthi CB, Singh RK. Proinflammatory IL-17 levels in serum/cerebrospinal fluid of patients with neurodegenerative diseases: a meta-analysis study. Naunyn Schmiedebergs Arch Pharmacol. 2023;396(3):577–588. doi:10.1007/S00210-022-02357-6
12. Huang J, Li X, Zhu Q, et al. Imbalance of Th17 cells, Treg cells and associated cytokines in patients with systemic lupus erythematosus: a meta-analysis. Front Immunol. 2024;15. doi:10.3389/FIMMU.2024.1425847
13. Ye X, Dai M, Xiang Z. Prognostic role of systemic inflammation response index in patients with non-small cell lung cancer: a meta-analysis. BMJ Open. 2024;14(11):e087841. doi:10.1136/BMJOPEN-2024-087841
14. World Health Organization. Noncommunicable diseases. Available from: https://www.who.int/news-room/fact-sheets/detail/noncommunicable-diseases.AccessedMarch20, 2025 .
15. Kannan G, Paul BM, Thangaraj P. Stimulation, regulation, and inflammaging interventions of natural compounds on nuclear factor kappa B (NF-kB) pathway: a comprehensive review. Inflammopharmacology. 2025;33(1):145–162. doi:10.1007/S10787-024-01635-4/METRICS
16. Tredicine M, Mucci M, Recchiuti A, et al. Immunoregulatory mechanisms of the arachidonic acid pathway in cancer. FEBS Lett. 2025;599:927–951. doi:10.1002/1873-3468.70013
17. Amedei A, Parolini C. Editorial: epigenetics of inflammatory reactions and pharmacological modulation. Front Pharmacol. 2024;15. doi:10.3389/FPHAR.2024.1505196
18. Parolini C. The role of marine n-3 polyunsaturated fatty acids in inflammatory-based disease: the case of rheumatoid arthritis. Mar Drugs. 2023;22(1):17. doi:10.3390/MD22010017
19. Mińczuk K, Baranowska‐Kuczko M, Krzyżewska A, et al. Cross-talk between the (Endo)Cannabinoid and renin-angiotensin systems: basic evidence and potential therapeutic significance. Int J mol Sci. 2022;23(11):6350. doi:10.3390/IJMS23116350
20. Lissoni P, Brivio F, Fumagalli L, et al. How to modulate the immunoinflammatory biological response in cancer, autoimmunity and viral infections by a psychoneuroimmune approach. Novel Approaches in Cancer Study. 2021;5. doi:10.31031/NACS.2021.05.000622
21. Barbé-Tuana F, Funchal G, Schmitz CRR, et al. The interplay between immunosenescence and age-related diseases. Semin Immunopathol. 2020;42(5):545–557. doi:10.1007/S00281-020-00806-Z
22. Franceschi C, Garagnani P, Parini P, et al. Inflammaging: a new immune-metabolic viewpoint for age-related diseases. Nat Rev Endocrinol. 2018;14(10):576–590. doi:10.1038/S41574-018-0059-4
23. World Health Organization. Ageing and health. Available from: https://www.who.int/news-room/fact-sheets/detail/ageing-and-health.AccessedMarch20, 2025 .
24. Bottaccioli AG, Bottaccioli F, Minelli A. Stress and the psyche-brain-immune network in psychiatric diseases based on psychoneuroendocrineimmunology: a concise review. Ann N Y Acad Sci. 2019;1437(1):31–42. doi:10.1111/NYAS.13728
25. Bottaccioli AG, Bologna M, Bottaccioli F. Psychic life-biological molecule bidirectional relationship: pathways, mechanisms, and consequences for medical and psychological sciences–a narrative review. Int J mol Sci. 2022;23(7):3932. doi:10.3390/IJMS23073932
26. McEwen BS, Akil H. Revisiting the stress concept: implications for affective disorders. J Neurosci. 2020;40(1):12–21. doi:10.1523/JNEUROSCI.0733-19.2019
27. Lissoni P, Rovelli F, Messina G, et al. Psycho-neuro-endocrine-immunology therapy of cancer, autoimmunity, geriatric disorders, Covid-19, and hypertension. Methods mol Biol. 2025;2868:111–132. doi:10.1007/978-1-0716-4200-9_7
28. Cardinali DP, Pandi-Perumal SR, Brown GM. Melatonin as a chronobiotic and cytoprotector in non-communicable diseases: more than an antioxidant. Subcell Biochem. 2024;217–244. doi:10.1007/978-3-031-66768-8_11
29. Vigano M, Wang L, As’sadiq A, et al. Impact of cannabinoids on cancer outcomes in patients receiving immune checkpoint inhibitor immunotherapy. Front Immunol. 2025;16. doi:10.3389/FIMMU.2025.1497829
30. Kunvariya AD, Dave SA, Modi ZJ, et al. Exploration of multifaceted molecular mechanism of angiotensin-converting enzyme 2 (ACE2) in pathogenesis of various diseases. Heliyon. 2023;9(5):e15644. doi:10.1016/J.HELIYON.2023.E15644
31. World Health Organization. WHO classification of tumours online. Available from: https://tumourclassification.iarc.who.int/welcome/.AccessedMarch20, 2025 .
32. Awwad ZM, El-Ganainy SO, ElMallah AI, et al. Assessment of pregabalin-induced cardiotoxicity in rats: mechanistic role of angiotensin 1-7. Cardiovasc Toxicol. 2020;20(3):301–311. doi:10.1007/S12012-019-09553-6
33. Maestroni GJM. The immunoneuroendocrine role of melatonin. J Pineal Res. 1993;14(1):1–10. doi:10.1111/J.1600-079X.1993.TB00478.X
34. Tshikhudo PP, Mabhaudhi T, Koorbanally NA, et al. Anticancer potential of β-carboline alkaloids: an updated mechanistic overview. Chem Biodivers. 2024;21(2). doi:10.1002/CBDV.202301263
35. McQuaid RJ, McInnis OA, Abizaid A, et al. Making room for oxytocin in understanding depression. Neurosci Biobehav Rev. 2014;45:305–322. doi:10.1016/J.NEUBIOREV.2014.07.005
36. Gu L, Li H, Chen L, et al. Prognostic role of lymphocyte to monocyte ratio for patients with cancer: evidence from a systematic review and meta-analysis. Oncotarget. 2016;7(22):31926–31942. doi:10.18632/ONCOTARGET.7876
37. Kose N, Akin F, Yildirim T, et al. The association between the lymphocyte-to-monocyte ratio and coronary artery disease severity in patients with stable coronary artery disease. Eur Rev Med Pharmacol Sci. 2019;23(6):2570–2575. doi:10.26355/EURREV_201903_17406
38. Lissoni P. How to modulate the immunoinflammatory biological response in cancer, autoimmunity and viral infections by a psychoneuroimmune approach. Nov Approaches Cancer Study. 2021;5(5). doi:10.31031/NACS.2021.05.000622
39. Tan DX, Reiter RJ. Mitochondria: the birth place, battle ground and the site of melatonin metabolism in cells. Melatonin Research. 2019;2(1):44–66. doi:10.32794/mr11250011
40. Rafiyian M, Reiter RJ, Rasooli Manesh SM, et al. Programmed cell death and melatonin: a comprehensive review. Funct Integr Genomics. 2024;24(5):169. doi:10.1007/S10142-024-01454-4
41. Reiter RJ, De Almeida Chuffa LG, Simão VA, et al. Melatonin and vitamin D as potential synergistic adjuvants for cancer therapy. Int J Oncol. 2024;65(6). doi:10.3892/IJO.2024.5702
42. Luo Z, Tang YY, Zhou L. Melatonin as an adjunctive therapy in cardiovascular disease management. Sci Prog. 2024;107(4). doi:10.1177/00368504241299993
43. Méndez N, Corvalan F, Halabi D, et al. From gestational chronodisruption to noncommunicable diseases: pathophysiological mechanisms of programming of adult diseases, and the potential therapeutic role of melatonin. J Pineal Res. 2023;75(4). doi:10.1111/JPI.12908
44. Cardinali DP. Melatonin: clinical perspectives in neurodegeneration. Front Endocrinol (Lausanne). 2019;10. doi:10.3389/fendo.2019.00480
45. Martinez-Ruiz L, López-Rodríguez A, Florido J, et al. Patient-derived tumor models in cancer research: evaluation of the oncostatic effects of melatonin. Biomed Pharmacother. 2023;167:115581. doi:10.1016/J.BIOPHA.2023.115581
46. Cruz-Sanabria F, Bruno S, Crippa A, et al. Optimizing the time and dose of melatonin as a sleep-promoting drug: a systematic review of randomized controlled trials and dose-response meta-analysis. J Pineal Res. 2024;76(5). doi:10.1111/JPI.12985
47. Miranda-Riestra A, Cercós MG, Trueta C, et al. Participation of Ca2+-calmodulin-dependent protein kinase II in the antidepressant-like effects of melatonin. Mol Pharmacol. 2024;106(3):107–116. doi:10.1124/MOLPHARM.124.000890
48. Thakur T, Saini V, Grover S, et al. Role of oral melatonin in prevention of postoperative delirium in patients undergoing elective surgery under general anesthesia: a randomized controlled trial. Indian J Psychiatry. 2024;66(5):457–462. doi:10.4103/INDIANJPSYCHIATRY.INDIANJPSYCHIATRY_965_23
49. Lissoni P, Messina G, Rovelli F. Cancer as the main aging factor for humans: the fundamental role of 5-methoxy-tryptamine in reversal of cancer-induced aging processes in metabolic and immune reactions by non-melatonin pineal hormones. Curr Aging Sci. 2012;5(3):231–235. doi:10.2174/1874609811205030010
50. Airaksinen MM, Kari I. β-carbolines, psychoactive compounds in the mammalian body. Part I: occurrence, origin and metabolism. Med Biol. 1981;59(1):21–34.
51. Airaksinen MM, Kari I. β-carbolines, psychoactive compounds in the mammalian body. Part II: effects. Med Biol. 1981;59(4):190–211.
52. Tapp E. The human pineal gland in malignancy. J Neural Transm. 1980;48(2):119–129. doi:10.1007/BF01250040
53. Gheblawi M, Wang K, Viveiros A, et al. Angiotensin-converting enzyme 2: SARS-CoV-2 receptor and regulator of the renin-angiotensin system: celebrating the 20th anniversary of the discovery of ACE2. Circ Res. 2020;126(10):1456–1474. doi:10.1161/CIRCRESAHA.120.317015
54. Shukla AK, Banerjee M. Angiotensin-converting-enzyme 2 and renin-angiotensin system inhibitors in COVID-19: an update. High Blood Pressure and Cardiovascular Prevention. 2021;28(2):129–139. doi:10.1007/s40292-021-00439-9
55. Shukla AK, Awasthi K, Usman K, et al. Role of renin-angiotensin system/angiotensin converting enzyme-2 mechanism and enhanced COVID-19 susceptibility in type 2 diabetes mellitus. World J Diabetes. 2024;15(4):606–622. doi:10.4239/WJD.V15.I4.606
56. Capettini L SA, Montecucco F, Mach F, et al. Role of renin-angiotensin system in inflammation, immunity and aging. Curr Pharm Des. 2012;18(7):963–970. doi:10.2174/138161212799436593
57. Platten M, Youssef S, Eun MH, et al. Blocking angiotensin-converting enzyme induces potent regulatory T cells and modulates TH1- and TH17-mediated autoimmunity. Proc Natl Acad Sci U S A. 2009;106(35):14948–14953. doi:10.1073/PNAS.0903958106
58. Du N, Feng J, Hu LJ, et al. Angiotensin II receptor type 1 blockers suppress the cell proliferation effects of angiotensin II in breast cancer cells by inhibiting AT1R signaling. Oncol Rep. 2012;27(6):1893–1903. doi:10.3892/or.2012.1720
59. Moazen S, Arjmand MH. The role of local angiotensin II/angiotensin type 1 receptor in endometriosis: a potential target for new treatment approaches. Curr Mol Pharmacol. 2024;17. doi:10.2174/0118761429315431240712100124
60. Thimm C, Adjaye J. Untangling the uncertain role of overactivation of the renin-angiotensin-aldosterone system with the aging process based on sodium wasting human models. Int J mol Sci. 2024;25(17):9332. doi:10.3390/IJMS25179332
61. Santos RAS, Sampaio WO, Alzamora AC, et al. The ACE2/angiotensin-(1-7)/Mas axis of the renin-angiotensin system: focus on angiotensin-(1-7). Physiol Rev. 2018;98(1):505–553. doi:10.1152/physrev.00023.2016
62. Rodrigues Prestes TR, Rocha NP, Miranda AS, et al. The anti-inflammatory potential of ACE2/Angiotensin-(1-7)/Mas receptor axis: evidence from basic and clinical research. Curr Drug Targets. 2017;18(11). doi:10.2174/1389450117666160727142401
63. Simões E Silva AC, Silveira KD, Ferreira AJ, et al. ACE2, angiotensin-(1-7) and Mas receptor axis in inflammation and fibrosis. Br J Pharmacol. 2013;169(3):477–492. doi:10.1111/bph.12159
64. Zheng J, Hao H. Targeting renal damage: the ACE2/Ang-(1-7)/mas axis in chronic kidney disease. Cell Signal. 2024;124:111413. doi:10.1016/J.CELLSIG.2024.111413
65. Mann SJ, Christos PJ. ACE inhibitors and ARBs: do they reduce the risk of cancer? J Clin Hypertens. 2014;16(1):6–7. doi:10.1111/JCH.12225
66. Rosenthal T, Gavras I. Renin-angiotensin inhibition in combating malignancy: a review. Anticancer Res. 2019;39(9):4597–4602. doi:10.21873/ANTICANRES.13639
67. Campos LA, Cipolla-Neto J, Amaral FG, et al. The Angiotensin-melatonin axis. Int J Hypertens. 2013;2013:1–7. doi:10.1155/2013/521783
68. Mompeón A, Lázaro-Franco M, Bueno-Betí C, et al. Estradiol, acting through ERα, induces endothelial non-classic renin-angiotensin system increasing angiotensin 1-7 production. mol Cell Endocrinol. 2016;422:1–8. doi:10.1016/J.MCE.2015.11.004
69. Shalata W, Abu Saleh O, Tourkey L, et al. The efficacy of cannabis in oncology patient care and its anti-tumor effects. Cancers. 2024;16(16):2909. doi:10.3390/CANCERS16162909
70. Lissoni P, Resentini M, Mauri R, et al. Effects of tetrahydrocannabinol on melatonin secretion in man. Horm Metab Res. 1986;18(1):77–78. doi:10.1055/S-2007-1012235
71. Koch M, Ferreirós N, Geisslinger G, et al. Rhythmic control of endocannabinoids in the rat pineal gland. Chronobiol Int. 2015;32(6):869–874. doi:10.3109/07420528.2015.1041596
72. Alenina N, Bader M. ACE2 in brain physiology and pathophysiology: evidence from transgenic animal models. Neurochem Res. 2019;44(6):1323–1329. doi:10.1007/S11064-018-2679-4
73. Li J, Kong X, Liu T, et al. The role of ACE2 in neurological disorders: from underlying mechanisms to the neurological impact of COVID-19. Int J mol Sci. 2024;25(18). doi:10.3390/IJMS25189960
74. Beurel E, Lowell JA. Th17 cells in depression. Brain Behav Immun. 2018;69:28–34. doi:10.1016/J.BBI.2017.08.001
75. Nunes-Souza V, Alenina N, Qadri F, et al. ACE2 knockout mice are resistant to high-fat diet-induced obesity in an age-dependent manner. Int J Mol Sci. 2024;25(17):9515. doi:10.3390/IJMS25179515
76. Imenshahidi M, Roohbakhsh A, Hosseinzadeh H. Effects of telmisartan on metabolic syndrome components: a comprehensive review. Biomed Pharmacother. 2024;171:116169. doi:10.1016/J.BIOPHA.2024.116169
Paolo Lissoni,1 Franco Rovelli,1 Alejandra Monzon,1 Giusy Messina,1 Nicoletta Merli,1 Rosanna Tartarelli,2 Simonetta Tassoni,2 Francesca Zecchinato,3 Ana Cristina Simoes-e-Silva,4 Agnese Valentini,5 Giuseppe Di Fede,1 Daniel P Cardinali6
1Institute of Biological Medicine/International Institute of PNEI, Milan, Italy; 2PNEI Psychological Service, Pietrasanta, Lucca, Italy; 3Biotherapy Service, Verona, Italy; 4Facultade de Medicina, Belo Horizonte, Brazil; 5Madonna del Soccorso Hospital, San Benedetto del Tronto, Ascoli-Piceno, Italy; 6Facultad de Ciencias Médicas, Pontificia Universidad Católica Argentina, Buenos Aires, Argentina
Correspondence: Daniel P Cardinali, Facultad de Ciencias Médicas, Pontificia Universidad Católica Argentina, Av Moreau de Justo 1300, Buenos Aires, C1107AFA, Argentina, Email [email protected]; [email protected]
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
© 2025. This work is licensed under https://creativecommons.org/licenses/by-nc/4.0/ (the “License”). Notwithstanding the ProQuest Terms and Conditions, you may use this content in accordance with the terms of the License.
Abstract
Purpose: Recent advancements in psycho-neuro-endocrine-immunology indicate that numerous noncommunicable diseases (NCDs) originate from disruptions in the cytokine immune network, resulting in chronic inflammatory responses. This persistent low-degree inflammation is attributed to deficiencies in crucial endogenous anti-inflammatory neuroendocrine systems, including the pineal gland, the endocannabinoid system, and the angiotensin-converting enzyme 2 / angiotensin 1– 7 axis. The administration of pineal methoxyindoles (melatonin, 5-methoxytryptamine), cannabinoids, and angiotensin 1– 7 may entail potential therapeutic benefits for NCDs, particularly for patients who do not respond to conventional treatments.
Patients and Methods: This study evaluates the safety and efficacy of a neuroimmune regimen comprising melatonin (100 mg/day at night), 5-methoxytryptamine (30 mg in the early afternoon), angiotensin 1– 7 (0.5 mg twice daily), and cannabidiol (20 mg twice daily) in 306 patients with NCDs, including advanced cancer, autoimmune diseases, neurodegenerative disorders, depression, and cardiovascular disease.
Results: The neuroimmune regimen successfully halted cancer progression in 68% of cancer patients, who also reported improvements in mood, sleep, and relief from anxiety, pain, and fatigue. In patients with autoimmune diseases, the treatment effectively controlled the disease process, remarkable in cases of multiple sclerosis. Additionally, positive outcomes were observed in patients with Parkinson’s disease, Alzheimer’s disease, and depression.
Conclusion: Randomized controlled trials are required to assess this therapeutic approach for NCDs that includes endogenous neuroendocrine molecules regulating immune responses in an anti-inflammatory manner.
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer