2,358
Views
7
CrossRef citations to date
0
Altmetric
Review

The role of the sex hormone-gut microbiome axis in tumor immunotherapy

, , , , , , , , , & show all
Article: 2185035 | Received 19 Oct 2022, Accepted 21 Feb 2023, Published online: 07 Mar 2023

ABSTRACT

Accumulating evidence suggested that both gut microbiome and sex play a critical role in the efficacy of immune checkpoint blockade therapy. Considering the reciprocal relationship between sex hormones and gut microbiome, the sex hormone-gut microbiome axis may participate in the regulation of the response to immune checkpoint inhibitors (ICIs). In this review, it was attempted to summarize the current knowledge about the influences of both sex and gut microbiome on the antitumor efficacy of ICIs and describe the interaction between sex hormones and gut microbiome. Accordingly, this review discussed the potential of enhancing the antitumor efficacy of ICIs through regulating the levels of sex hormones through manipulation of gut microbiome. Collectively, this review provided reliable evidence concerning the role of the sex hormone-gut microbiome axis in tumor immunotherapy.

Introduction

Through unleashing of antitumor T cell activity, immune checkpoint inhibitors (ICIs) have achieved great success in diverse types of cancer. However, some cancer patients have not responded to ICIs, indicating the necessity of searching for the reasons for the low responsiveness to ICIsCitation1. Among the multiple potential factors, certain gut microbiome signatures have been found to be associated with a higher response to ICIsCitation2–4. Both gut microbes and bacterial-derived metabolites may contribute to the influences of gut microbiome on the outcomes after receiving ICIs; however, the underlying mechanisms have not yet been fully clarifiedCitation5.

Emerging evidence indicated the existence of the sex hormone-gut microbiome axis. The sex-based differences in gut microbiome have been extensively investigated. Changes in sex hormone levels induced by gonadectomyCitation6, menstrual cycle and menopausal statusCitation7,Citation8, or certain diseases (e.g., polycystic ovary syndrome (PCOS))Citation9 can alter the gut microbial composition. In turn, transfer of gut microbiotaCitation10 or treatment with antibioticsCitation11 resulted in significant changes in levels of sex hormones. The interaction between sex hormones and gut microbiome, termed the sex hormone-gut microbiome axis here, plays a vital role in multiple diseases, such as PCOSCitation9, cardiovascular diseaseCitation12, mental disordersCitation13, etc.

Clinical meta-analysis indicated that the magnitude of benefit derived from ICIs is largely sex-dependentCitation14,Citation15. Moreover, sex hormone receptors were recently identified to play an important role in the sex-based immune response to ICIs via regulating CD8+ T cell functionCitation16–19. Considering the existence of the sex hormone-gut microbiome axis, gut microbiome may affect the antitumor efficacy of ICIs by regulating the host’s sex hormone levels. The present study aimed to review recent advances in understanding the underlying mechanisms by which gut microbiome and sex hormones could affect the immune response to ICIs. In particular, the interaction mechanisms between sex hormones and gut microbiome were explored, and the possibility of enhancing antitumor efficacy of ICIs via manipulating the sex hormone-gut microbiome axis was assessed.

Gut microbiome could modulate the response to ICIs

The effects of gut microbiome on antitumor immunity were first revealed in mice with disruption of gut mucosal integrity induced by total body irradiation or cyclophosphamideCitation20,Citation21. The translocation of certain gut microbes, such as Lactobacillus and Bifidobacterium, into secondary lymphoid organs triggered by gut mucosal disruption could promote the activation of CD4+ or CD8+ T cells and improve the antitumor outcomes. Meanwhile, it was demonstrated that gut microbiome was necessary for the efficacy of cytosine-phosphate-guanine (CpG)-oligonucleotide-based tumor immunotherapyCitation22. Then, the essential role of gut microbiome in ICI immunotherapy was investigated in anti-cytotoxic T-lymphocyte-associated protein 4 (anti-CTLA-4) or anti-programmed death-ligand 1 (anti-PD-L1) antibody-treated mice and the relevant bacterial species were identified by 16S ribosomal RNA sequencingCitation23,Citation24. It was found that the antitumor effects of CTLA-4 blockade depended on specific Bacteroides speciesCitation24, while oral administration of Bifidobacterium facilitated anti-PD-L1 efficacy against melanomaCitation4. These valuable findings from preclinical mouse models suggested a critical role of gut microbiome in cancer immunotherapy and inspired further investigation on human cancer patients. In 2018, three independent studies demonstrated that intestinal microbiome profoundly affected responses to anti-programmed cell death protein 1 (anti-PD-1)/anti-PD-L1 immunotherapy in patients with melanoma, non-small cell lung cancer (NSCLC), and renal cell carcinomaCitation2–4. However, no consensus microbial signals associated with favorable response to ICIs were identified in these three studies, suggesting that function, rather than specific species, may better indicate the effects of intestinal microbiotaCitation25. Due to the limited concordance among different studies and the lack of understanding of the precise composition of a favorable gut microbiome, fecal microbiota transplantation (FMT) exhibited to be a promising method to eliminate resistance to ICI immunotherapyCitation26,Citation27. Multiple clinical trials have assessed the feasibility of improving the efficacy and safety of ICIs through FMTCitation28,Citation29. Two clinical trials have concentrated on the use of FMT to improve ICI response in patients with melanoma (Identifiers: NCT03341143 and NCT04988841), and one trial for metastatic castration-resistant prostate cancer (Identifier: NCT04116775) has moved to a phase II trial.

The influences of gut microbiome on the antitumor efficacy of ICIs may be mediated by multiple mechanisms (), which have not yet been fully exploredCitation5. Both gut microbes and their metabolites may contribute to the effects of gut microbiome on the cancer immune response. For instance, bacterial immunostimulants, such as peptidoglycanCitation30, polysaccharideCitation24, exopolysaccharideCitation31, and type 1 fimbriae adhesion portionCitation32, may enhance the antitumor effects of ICIs through strengthening both innate immunity and T cell functions in mice. Bacteria penetrating the mucus and submucosal lymphoid organsCitation21 or even the tumor sitesCitation33 may also stimulate the antitumor immune response in mice. Besides, a cross-molecular mimicry may exist between bacteria and tumor-associated antigens, leading to the induction of cross-reactive CD8+ T cell responsesCitation34. Inspired by the similarity of gut microbiota and tumor-derived antigens, a therapeutic vaccine has been developed using “oncomimic” peptides in conjunction with anti-PD-1 for treatment of colorectal cancer (Identifier: NCT05350501).

Table 1. Possible mechanisms of the influences of gut microbiome on the antitumor efficacy of ICIs.

In addition to the immunomodulatory microbial components, the effects of gut microbiome on the antitumor immune response to ICIs can be mediated by their metabolites. Short-chain fatty acidsCitation35–37, the major end-products of gut microbiota-derived metabolites, and some short-chain fatty acid-producing bacterial species, such as Akkermansia muciniphila Citation3, Lachnospiraceae Citation38, and Lactobacillus Citation39, have been found to be associated with favorable response to ICIs in cancer patients. Particularly, butyrate could directly enhance the antitumor cytotoxic CD8+ T cell responses by promoting IL-12 signaling pathway and increase the efficacy of anti-PD-L1 therapy in miceCitation40. Similarly, the purine metabolite inosine produced by certain bacteria, such as Bifidobacterium pseudolongum and Akkermansia muciniphila, could improve response to ICIs in mouse models of cancer via activating anti-tumor T cells through adenosine A2A receptorCitation41. Besides, inosine could serve as an alternative carbon source for CD8+ T cell function under glucose restriction and relieve tumor-imposed metabolic restrictions on T cellsCitation42. Supplementation with inosine enhanced the anti-tumor efficacy of ICIs in miceCitation41,Citation42. Another microbial metabolite correlated with efficacy of ICIs is trimethylamine oxide (TMAO)Citation43. Choline or carnitine in the food can be transformed into the precursor trimethylamine by gut microbiome, which may be catalyzed to generate TMAO in the liver. Either intratumoral injection of TMAO or oral supplement with choline enhanced antitumor activity of anti-PD-1 in mice with triple-negative breast cancerCitation43. Other gut microbiota-derived metabolites, such as anacardic acidsCitation44 and secondary bile acidsCitation45, may also affect the antitumor effects of ICIs. However, it is noteworthy that both TMAO and secondary bile acids may also promote carcinogenesis, especially in the colonCitation46–48. In addition, higher plasma levels of TMAO and precursors may increase the risk of coronary heart diseaseCitation49.

Some microbial metabolites may also decrease the antitumor efficacy of ICIs. For instance, the tryptophan released from degradation of dietary proteins can be converted into various metabolites by gut microbiome, such as indole, indole-related compounds, and kynurenine. As a typical immunosuppressive tryptophan metabolite, a higher kynurenine/tryptophan ratio has been found to be associated with a poor response to anti-PD-1 in NSCLC patientsCitation50. Moreover, the influences of microbial metabolites on anti-tumor immunity could be largely situation-dependent. For instance, high blood butyrate and propionate levels were reported to be associated with resistance to CTLA-4 blockade in patients with metastatic melanoma, and oral administration of sodium butyrate diminished antitumor efficacy of anti-CTLA-4 in miceCitation51. It could be attributed to the difference between anti-CTLA-4 and anti-PD-1/PD-L1, which needs to be validated in the future. In addition, considering the large pool size of gut microbiota-derived metabolites, the causal relationship for most of the metabolites should be further confirmed, although a strong correlation may be identified.

Of note, ICI immunotherapy can also alter the composition of the gut microbiome. It has been shown that there is a variation in the gut microbiome composition during anti-PD-1 immunotherapy in patients with hepatocellular carcinomaCitation38. The effects of ICIs on gut microbiome were also observed in an animal studyCitation24. Furthermore, our previous study indicated that anti-PD-L1 could significantly alter the composition of the gut microbiome and decrease the relative abundance of Lachnospiraceae in female mice while exerting no effect on male miceCitation52. The underlying mechanisms of the impact of ICIs on gut microbiome have yet to be comprehensively clarified. The activation of T cells in the intestine could be associated with these effectsCitation24.

Sex-based differences in response to ICIs

Various factors have been found to be associated with antitumor response to immunotherapy, including tumor-cell-intrinsic features (e.g., PD-L1 expression, tumor-associated antigens, tumor burden, tumor mutational burden, mismatch repair deficiency, and epigenetic alterations), tumor immune microenvironment signatures (e.g., tumor-infiltration lymphocytes and the presence of immunosuppressive cells), and environmental factors, such as diets and intake of antibioticsCitation1,Citation53–55. However, recognition of the importance of sex on ICI response is relatively recent. Sex-based differences in tumor incidence and mortality are evident for most types of cancer, in which male patients have higher incidence rates ranging from 1.26:1 to 4.86:1Citation56. These differences may be partially attributed to sex-based differences in the immune system. Generally, women have stronger innate and adaptive immunity, higher incidence rates of autoimmune diseases, better vaccine responses, and greater tolerability of adverse effects of vaccinationCitation57,Citation58. Evidence from meta-analysis of clinical trials mainly indicated that men responded better to ICIs than womenCitation14,Citation59–62. However, this trend has not always been confirmed. A trend of higher response rate in female patients compared with male patients with NSCLC was also reportedCitation63, which is contradictory to other studiesCitation14,Citation60,Citation61. In contrast, one meta-analysis included 23 randomized clinical trials (9322 men and 4399 women) demonstrated no statistically significant difference in response to ICI immunotherapy between the sexesCitation64. Moreover, female patients with advanced lung cancer achieved a significantly greater benefit from the combined therapy of chemotherapy and anti-PD-1/PD-L1, whereas male patients responded better to anti-PD-1 aloneCitation15. Thus, addressing these concerns by meta-analysis pooling different clinical trials may not be adequateCitation63. The relatively large fluctuation of levels of sex hormones among patients due to either physiological or pathological factors may partially account for the discrepancy. Besides, our previous studies on mice revealed that anti-PD-L1 treatment could significantly affect male sex hormone levels, further complicating our understanding of the role of sex in tumor immunotherapy. Notwithstanding, significantly higher tumor mutational burden, single-nucleotide variation neoantigen load, and PD-L1 expression level could be found in male patients with melanomaCitation63,Citation65. In addition, studies showed that androgen deprivation therapy could significantly enhance the antitumor efficacy of ICIs in miceCitation52,Citation66. Although whether there is a sex-based difference in survival benefits from ICIs remains controversial, which may be addressed in future clinical studies covering more types of cancer other than melanoma and lung cancer, the effects of sex-relevant features on the antitumor immunotherapy deserve oncologists’ attention.

Many factors including hormones, genetic differences, and environmental factors are involved in the formation of sex disparitiesCitation67,Citation68. Although sex hormones cannot account for all sex-based differences in cancerCitation56, they play a critical role in anticancer immunityCitation68. Sex hormone receptors are nearly expressed in all immune cells, which participate in the regulation of the expression levels of many immune-related genesCitation68. In general, estrogen enhances both innate and adaptive immunity, while androgen suppresses immune cell activityCitation58. However, most studies demonstrated that male patients tend to achieve greater survival benefits from ICI immunotherapy, which seems to be inconsistent with the effects of sex hormones. The sex-based immune features, such as tumor mutational burden and tumor infiltration of immune cells, may contribute to the differences in the efficiency of immunotherapy between male and female patientsCitation63. On the other hand, our previous study showed that anti-PD-L1 could significantly downregulate the levels of sex hormones in male mice rather than in female mice, which enhanced the antitumor efficacy of anti-PD-L1Citation52. In contrast, Tulchiner et al. reported that anti-PD-1 immunotherapy significantly increased estradiol and luteinizing hormone (LH)/follicle-stimulating hormone (FSH) ratio in male patients with metastatic renal cell carcinoma from the beginning of therapy to week 12 of follow-up, while it had no influence on testosterone level in both sexesCitation69. The potential effects of ICIs on the levels of sex hormones and in turn their impacts on the sex-based differences in the response to ICIs have not yet been fully explored and deserve further investigation.

Sex-based differences in gut microbiome

A growing body of evidence from both human and animal studies indicated the existence of sex-based differences in microbiota compositionCitation70–74. Generally, women tend to have a higher alpha diversity. In a study of 1135 individuals from a population-based Netherlands cohort, women had a greater microbial diversity based on the values of the Shannon indexCitation6. Similarly, one study involving 551 healthy Chinese participants also revealed that women had a significantly higher alpha diversity in the fecal microbiota as assessed by the observed number of operational taxonomic units and the values of the Shannon indexCitation75. In another study from Italy, a significant increase in the Chao1 index and Shannon index was found in the mucosa-associated microbiota of female participants sampled by sigmoid brushCitation76. Besides, a significantly higher species richness estimated by the Chao1 index was also found in wild-type female mice compared to male miceCitation73. Firmicutes and Bacteroidetes are two of the most primary bacteria identified in the gut of both humans and animals, and the ratio of Firmicutes to Bacteroidetes (F/B ratio) is a widely used marker for gut dysbiosis, which has been associated with a number of health status-related factors, such as hypertensionCitation77 and obesityCitation78. The F/B ratio also tends to be higher in women. For instance, one recent study from Ukraine involving 2301 healthy participants revealed that the F/B ratio was significantly higher in women than in menCitation79. A higher F/B ratio was also found in healthy post-menopausal women than in pre-menopausal women or their corresponding age-matched menCitation7. In addition, a lower abundance of Bacteroidetes was identified in women from a cohort conducted in the United StatesCitation80. Notably, the alpha diversity of gut microbiome has been reported to be strongly negatively correlated with the relative abundance of Bacteroidetes Citation81,Citation82. The higher microbial alpha diversity in females may be attributed to their relatively lower abundance of Bacteroidetes. However, the F/B ratio significantly varies among individuals and is noticeably affected by geographical latitudeCitation82,Citation83, and no significant effect of sex on the F/B ratio has been found in some studies, possibly due to the large geographic scale or limited sample sizeCitation83,Citation84. Besides, it is noteworthy that there are also sex-based differences in the gut microbiome in cancer patientsCitation85. Moreover, there are distinct changes in microbial alpha diversity and community composition between the sexes during the development of colorectal cancerCitation85. These sex-based differences in gut microbiome have been suggested to contribute to the sex-based disparity in liver carcinogenesis in miceCitation48. The role of the sex-based differences in gut microbiome during cancer development and progression deserves further assessment.

Although summarizing the sex-based differentially abundant bacterial taxa is a challenge due to the lack of consistency among studies, some sex hormones-associated bacteria have been identifiedCitation86,Citation87. For instance, total levels of urinary estrogen in men and postmenopausal women were significantly associated with fecal Clostridia Citation88. The ratio of urinary estrogen metabolites to parent estrogen (estrone and estradiol) was positively correlated with the relative abundance of Clostridiales in healthy postmenopausal women, while it was inversely associated with the genus Bacteroides Citation89. The abundance of Acinetobacter, Dorea, Ruminococcus, and Megamonas was significantly positively correlated with serum testosterone levels in men, while the abundance of Slackia and Butyricimonas was negatively correlated with serum estradiol levels in womenCitation84. In addition, studies demonstrated that sex hormones play an essential role in shaping the host gut microbiome. Changes in levels of sex hormones induced by drug administration (e.g., oral contraceptives)Citation8, gonadectomyCitation6, menstrual cycle, and menopausal statusCitation7,Citation8, or certain diseases, such as polycystic ovary syndromeCitation9, could alter the diversity or composition of gut microbiome. Animal studies further confirmed the effects of sex hormones on gut microbiomeCitation90–93.

Although the underlying mechanisms remain elusive, several potential pathways may be involved in the regulation of gut microbiome by sex hormones. Firstly, bacteria that express β-glucuronidase enzyme can release free sex hormone molecules from the conjugated metabolites that previously formed in the liver, which may result in the liberation of the glucuronic acid group and produce energy for gut bacteriaCitation9. Secondly, as mentioned earlier, sex hormone receptors are widely expressed in immune cellsCitation68; thus, sex hormones may influence gut microbiome by regulating intestinal immune response. Knock-out of estrogen receptor β (ERβ) in female mice could result in significant changes in the composition of gut microbiomeCitation92. Besides, animal studies revealed substantial sex-based differences in IgA levelCitation71 and the expression levels of anti-microbial peptidesCitation94. These findings demonstrated that intestinal immunity might contribute to the regulation of sex hormones on gut microbiome. More recently, Nuriel-Ohayon et al. reported that progesterone supplementation increases the abundance of Bifidobacterium in mice and in vitro Citation95, indicating the favorable metabolism of sex hormones by certain bacterial taxa may also alter gut microbial composition.

Gut microbiome could participate in regulation of sex hormone levels

The concept of “microgenderome” has been used to describe the bidirectional interaction between sex hormones and gut microbiomeCitation96. Sex-specific microbiome profiles that emerged after puberty contribute to the levels of sex hormones, which affect the development of autoimmune disease in the non-obese diabetic mouse model of type 1 diabetes (T1D)Citation10. Elevating testosterone levels by transferring gut microbiota from adult male mice to immature female mice reduced T1D incidence. Furthermore, only particular microbes, such as segmented filamentous bacteria and a proteobacterium isolated from male mice, have been correlated with serum testosterone concentration and colonization with these bacteria conferred the protection against T1D in male miceCitation97. These findings indicate that some gut microbes can participate in the regulation of the levels of sex hormones and ultimately modify the host’s autoimmunity.

Several potential mechanisms may be involved in the regulation of the host’s levels of sex hormones by gut microbiome (). Firstly, gut microbes may participate in the metabolism of sex hormones in intestine through expressing certain enzymes. After glucuronidation mainly in the liver, the glucuronidated steroid sex hormones are excreted in urine or via biliary excretion to intestineCitation98. These conjugated sex hormones are re-absorbed via enterohepatic circulation after deconjugation by β-glucuronidase from intestinal bacteria. It has been found that there is a noticeable free androgen in the intestine, which is 70-fold higher than that in the serum of young adult menCitation98. Thus, gut microbiome may play an important role in regulating circulating androgen or estrogen levels through deconjugation by β-glucuronidase in intestineCitation98,Citation99. Totally, 279 unique microbiome-encoded β-glucuronidase proteins clustered into six unique structural categories have been identified in the Human Microbiome Project databaseCitation100, of which certain members within three classes could reactivate estrogens from their inactive glucuronide formsCitation101. Similarly, bacterial β-glucosidases and sulfatases may also participate in the deconjugation of conjugated sex hormone metabolites in intestineCitation87,Citation102. Importantly, human fecal bacteria can carry out a variety of reductive, oxidative, and hydrolytic reactions of androgens and estrogensCitation102. The typical steroid-metabolizing enzymes involved in bacterial metabolism of sex hormones include hydroxysteroid dehydrogenase (HSD) and steroid reductaseCitation103. For instance, Streptococcus mutans, which showed a higher abundance in the gut microbiome from steroid inhaler users in patients with irritable bowel syndromeCitation104, has been identified with the potential of metabolizing progesterone and testosterone by expressing 5α- and 5β-steroid reductases and 3α-, 17β-, and 20α-HSDsCitation105. More recently, evidence supported the existence of bacterial enzymes (with a high sequence homology to human 1720 lyase) that would be responsible for the androgenic steroid biosynthesis in specific intestine microbiota species, such as Ruminococcus gnavus, contributing to endocrine resistance in castration-resistant prostate cancerCitation11.

Figure 1. Potential mechanisms by which gut microbiome may participate in regulating the host’s sex hormone levels. (A) the glucuronidation of sex hormones catalyzed by uridine diphosphate-glucuronosyltransferase 2B (UGT2B) in the liver increases the water solubility, which promotes the excretion of the glucuronidated compounds via urine or bile to the small intestine. Part of the conjugated sex hormones are de-conjugated by β-glucuronidase from the commensal gut bacteria. After deconjugation, the free sex hormone molecules are reabsorbed via the portal system. (B) Certain bacterial enzymes, such as 3α-HSD, 17β-HSD, 20α-HSD, 5α-reductase, and 1720 lyase, may participate in the biosynthesis of steroid hormones in the intestine, whereas further investigation is required. (C) Some bacteria are important for the maintenance of the protective function of the mucus. Destruction of the intestinal barrier may facilitate the passage of gut bacteria into the systemic circulation and elicit a chronic state of inflammation, which may impair testicular function, including the testosterone production by Leydig cells. (D) Some gut microbes are involved in the metabolism of neuroactive compounds or regulation of the gut-brain mediator secretion, which may influence the activity of the central nervous system via the gut–brain axis. Gut microbiome may thus influence endogenous production of sex hormones via the hypothalamic–pituitary–gonadal axis.

Figure 1. Potential mechanisms by which gut microbiome may participate in regulating the host’s sex hormone levels. (A) the glucuronidation of sex hormones catalyzed by uridine diphosphate-glucuronosyltransferase 2B (UGT2B) in the liver increases the water solubility, which promotes the excretion of the glucuronidated compounds via urine or bile to the small intestine. Part of the conjugated sex hormones are de-conjugated by β-glucuronidase from the commensal gut bacteria. After deconjugation, the free sex hormone molecules are reabsorbed via the portal system. (B) Certain bacterial enzymes, such as 3α-HSD, 17β-HSD, 20α-HSD, 5α-reductase, and 1720 lyase, may participate in the biosynthesis of steroid hormones in the intestine, whereas further investigation is required. (C) Some bacteria are important for the maintenance of the protective function of the mucus. Destruction of the intestinal barrier may facilitate the passage of gut bacteria into the systemic circulation and elicit a chronic state of inflammation, which may impair testicular function, including the testosterone production by Leydig cells. (D) Some gut microbes are involved in the metabolism of neuroactive compounds or regulation of the gut-brain mediator secretion, which may influence the activity of the central nervous system via the gut–brain axis. Gut microbiome may thus influence endogenous production of sex hormones via the hypothalamic–pituitary–gonadal axis.

Secondly, gut microbes may also directly affect gonadal function. Some mucus-degrading bacteria (e.g., A. muciniphila) are essential for the maintenance of the protective function of the mucusCitation106,Citation107. Destruction of the intestinal mucosal barrier may facilitate translocation of gut bacteria from gut lumen into circulation, triggering systemic inflammation that inhibits production of testosterone by Leydig cellsCitation108. In healthy men, low-dose endotoxin challenge produced an acute systemic inflammatory response, followed by a significant decline in plasma testosterone levels, without influencing LH or FSHCitation109. Similarly, our previous findings revealed that anti-PD-L1 could significantly downregulate the testosterone levels in male mice without affecting LH or FSH levels, probably through inducing local or systemic inflammatory responseCitation52. Moreover, oral supplementation with Lactobacillus could significantly reduce diethylhexyl phthalate-induced increase in the serum lipopolysaccharide level and recover the testosterone concentration in male miceCitation110. All these evidences support that gut microbiome may participate in the regulation of the testis testosterone secretion via regulating inflammation. In addition, the microbiota-derived gut-brain mediators may influence gonadal hormone secretion through the gut-brain axis. In PCOS patients, oral administration of probiotic Bifidobacterium lactis V9 significantly promoted the secretion of gut-brain mediators, including ghrelin and peptide YY, decreased the ratio of LH/FSH, and increased the levels of sex hormones, demonstrating the possible involvement of the gut-brain axisCitation111. Collectively, gut microbiome may take part in regulation of the host’s sex hormone levels via a variety of pathways, in which the detailed mechanisms have not yet been fully explored.

Modulating levels of sex hormones via manipulation of gut microbiome to enhance antitumor efficacy of ICIs

Emerging evidence suggests that the sex hormone receptor signaling pathway is involved in modulation of CD8+ T cell function. Androgen receptor (AR) signaling could promote the transition from stem cell-like CD8+ T cells to terminally exhausted CD8+ T cells in male mice, and it was correlated with tumor-infiltrating CD8+ T cell exhaustion in cancer patientsCitation16. ERβ augmented the downstream TCR signaling cascade, and the combined use of ERβ agonist and anti-PD-1 substantially increased tumor-infiltrating CD8+ T cells and sensitized various syngeneic tumors to ICI immunotherapy in miceCitation18. In contrast, ERα could promote macrophage polarization toward an immune-suppressive state, leading to CD8+ T cell dysfunction and exhaustionCitation19. Inhibition of ERα using the selective estrogen receptor significantly increased the antitumor efficacy of ICIs in mouse models of melanomaCitation19. Taken together, it is a feasible approach to improve the response to ICI therapy through regulating the sex hormone receptor signaling pathway.

To date, trials that investigated the potency of the possible combination of sex hormone intervention and tumor immunotherapy mainly concentrated on treatment of prostate cancer and breast cancerCitation112. For instance, the combination of enzalutamide and a cancer vaccine significantly improved the overall survival rate in the TRAMP mouse spontaneous prostate cancer modelCitation113. In the semi – hormone-dependent Myc-CaP mouse tumor model, combining CpG and surgical orchiectomy or abiraterone reduced tumor burden and more effectively delayed tumor relapse than either single treatmentCitation66. Guan et al. reported that the AR expressed on CD8+ T cells could repress IFN-γ expression level and mediate the resistance to ICI therapyCitation17. Surgical orchiectomy plus enzalutamide with anti-PD-L1 antibodies led to significant tumor regression and increased the overall survival rate in either an androgen deprivation therapy plus anti-PD-1-resistant mouse prostate tumor model or an AR-negative mouse sarcoma tumor modelCitation17. Besides, multiple ongoing clinical trials are investigating the efficiency of ICIs in combination with anti-estrogen therapy for breast cancerCitation114. More recently, it was found that a combination of 17β-estradiol and anti-PD-L1 significantly inhibited MC38 colon tumor growth in male miceCitation115. The findings mentioned above confirmed the feasibility of improvement of the efficacy of ICIs by modulating the levels of sex hormones. However, it is noteworthy that most existing trials have concentrated on improving the treatment of sex hormone-dependent cancer. It is essential to clarify whether the combination therapy is superior to ICI monotherapy for non-sex hormone-dependent cancer considering the potential side effects of sex hormone therapy. Furthermore, some nonsteroidal AR antagonists, such as flutamide and enzalutamide, have been shown to inhibit early-phase T cell activation and suppress the antitumor efficacy of anti-PD-L1 in miceCitation66.

Recently, Pernigoni et al. reported that the commensal gut microbiome contributes to endocrine resistance in castration-resistant prostate cancer by providing an alternative source of androgensCitation11. Importantly, ablation of the gut microbiome with a cocktail of broad-spectrum antibiotics delayed the emergence of castration resistance in both TRAMP-C1 allograft and the Ptenpc–/– prostate conditional mouse modelsCitation11. These results provide a novel approach to regulate the levels of sex hormones via manipulation of gut microbiome, which may be appropriate for the adjuvant therapy of non-sex hormone-dependent cancer. Given the gut microbiome’s great volatility, manipulating gut microbiome’s function with antibiotics rather than maintaining a specific bacterial species composition is a more reliable approach. However, numerous clinical studies have shown a detrimental effect of broad-spectrum antibiotics on ICI therapy, while the underlying mechanisms have been poorly exploredCitation53. In contrast, our previous study showed that oral administration of colistin, a narrow-spectrum antibiotic, could significantly downregulate the testosterone level in male mice and enhance the antitumor efficacy of anti-PD-L1 antibodiesCitation52. These findings confirmed the feasibility of improving the efficacy of antitumor immunotherapy by modulating levels of sex hormones via manipulation of the gut microbiome with narrow-spectrum antibiotics (). Other commonly used methods to modulate gut microbiome, including FMT, probiotics, and prebiotics have also been tested for improving antitumor efficiency of ICI immunotherapyCitation26,Citation27,Citation116–118, which can also affect the host’s levels of sex hormonesCitation10,Citation111,Citation119. However, it remains elusive whether sex hormones play a role in this process. Besides, additional studies are needed to clarify whether there is a synergistic effect of sex hormones and other microbial metabolites on ICI therapy.

Figure 2. Modulating the levels of sex hormones via manipulating gut microbiome to facilitate antitumor efficacy of ICIs. Drugs, such as antibiotics, can alter the composition of gut microbiome and induce changes in the host’s sex hormone levels by interfering with their microbial metabolism in the intestine or regulating the endogenous production via the gut-brain axis or inflammation. Activation of AR or ERα signaling pathway may attenuate the antitumor efficacy of ICIs by inducing CD8+ T cell exhaustion, while ERβ may augment the downstream TCR signaling cascade and increase tumor-infiltrating CD8+ T cells. Thus, the antitumor efficacy of ICIs may be improved using drugs, such as antibiotics, to alter the gut microbiome to regulate the host’s levels of sex hormones. However, it is noteworthy that administration of broad-spectrum antibiotics may impair the antitumor efficacy of ICIs.

Figure 2. Modulating the levels of sex hormones via manipulating gut microbiome to facilitate antitumor efficacy of ICIs. Drugs, such as antibiotics, can alter the composition of gut microbiome and induce changes in the host’s sex hormone levels by interfering with their microbial metabolism in the intestine or regulating the endogenous production via the gut-brain axis or inflammation. Activation of AR or ERα signaling pathway may attenuate the antitumor efficacy of ICIs by inducing CD8+ T cell exhaustion, while ERβ may augment the downstream TCR signaling cascade and increase tumor-infiltrating CD8+ T cells. Thus, the antitumor efficacy of ICIs may be improved using drugs, such as antibiotics, to alter the gut microbiome to regulate the host’s levels of sex hormones. However, it is noteworthy that administration of broad-spectrum antibiotics may impair the antitumor efficacy of ICIs.

Conclusions

In conclusion, a growing body of evidence demonstrated that the sex hormone-gut microbiome axis might be involved in regulating the antitumor efficacy of ICIs (). However, the underlying mechanisms have not yet been fully elucidated. Moreover, the potential effects of ICIs on the patient’s levels of sex hormones and the gut microbiome further complicate the role of the sex hormone-gut microbiome axis in ICI immunotherapy. Further research is required to elucidate the underlying mechanisms of the gut microbiome-mediated regulation of levels of sex hormones and develop new methods to manipulate the levels of sex hormones via targeting gut microbiome. Besides, the importance of the sex hormone-gut microbiome axis in tumor immunotherapy should be further evaluated considering the fact that several other gut microbial metabolites have also been found to be associated with the patient’s response to ICIs. Altogether, the sex hormone-gut microbiome axis provides a promising target for improving antitumor efficacy of ICI immunotherapy.

Figure 3. The role of the sex hormone-gut microbiome axis in tumor immunotherapy. Gut microbiome can influence the host’s levels of sex hormones through either metabolizing sex hormones or regulating gonadal secretion. In turn, the sex hormones can alter the gut microbiome by either serving as an energy source to support the growth of certain bacteria or regulating intestinal immune homeostasis. The interaction between sex hormones and gut microbiome constitutes the sex hormone-gut microbiome axis. Thus, the influences of gut microbiome and sex hormones on the patient’s response to ICIs can be simultaneously studied, although a growing body of evidence showed the effect of each of them on the antitumor efficacy of ICIs.

Figure 3. The role of the sex hormone-gut microbiome axis in tumor immunotherapy. Gut microbiome can influence the host’s levels of sex hormones through either metabolizing sex hormones or regulating gonadal secretion. In turn, the sex hormones can alter the gut microbiome by either serving as an energy source to support the growth of certain bacteria or regulating intestinal immune homeostasis. The interaction between sex hormones and gut microbiome constitutes the sex hormone-gut microbiome axis. Thus, the influences of gut microbiome and sex hormones on the patient’s response to ICIs can be simultaneously studied, although a growing body of evidence showed the effect of each of them on the antitumor efficacy of ICIs.

Author contributions

LW plotted the figures and wrote the manuscript. LT, DZ, MS, WL, SX, SJ, MZ, HM, and JL participated in drafting the manuscript. YW and ZB reviewed/edited the manuscript before submission.

Disclosure statement

No potential conflict of interest was reported by the author(s).

Data availability statement

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

Additional information

Funding

This study was supported by the National Natural Science Foundation of China (Grant No. 82101925) and the Natural Science Foundation of Shandong Province (Grant No. ZR2021QC215)

References

  • Dall’olio FG, Marabelle A, Caramella C, Garcia C, Aldea M, Chaput N, Robert C, Besse B. Tumour burden and efficacy of immune-checkpoint inhibitors. Nat Rev Clin Oncol. 2022;19(2):75–15. doi:10.1038/s41571-021-00564-3.
  • Matson V, Fessler J, Bao R, Chongsuwat T, Zha Y, Alegre ML, Luke JJ, Gajewski TF. The commensal microbiome is associated with anti–pd-1 efficacy in metastatic melanoma patients. Science. 2018;359(6371):104–108. doi:10.1126/science.aao3290.
  • Routy B, Le Chatelier E, Derosa L, Duong CPM, Alou MT, Daillere R, Fluckiger A, Messaoudene M, Rauber C, Roberti MP, et al. Gut microbiome influences efficacy of PD-1–based immunotherapy against epithelial tumors. Science. 2018;359(6371):91–97. doi:10.1126/science.aan3706.
  • Gopalakrishnan V, Spencer CN, Nezi L, Reuben A, Andrews MC, Karpinets TV, Prieto PA, Vicente D, Hoffman K, Wei SC, et al. Gut microbiome modulates response to anti–pd-1 immunotherapy in melanoma patients. Science. 2018;359(6371):97–103. doi:10.1126/science.aan4236.
  • Zhou CB, Zhou YL, Fang JY. Gut microbiota in cancer immune response and immunotherapy. Trends Cancer. 2021;7(7):647–660. doi:10.1016/j.trecan.2021.01.010.
  • Sinha T, Vich Vila A, Garmaeva S, Jankipersadsing SA, Imhann F, Collij V, Bonder MJ, Jiang X, Gurry T, Alm EJ, et al. Analysis of 1135 gut metagenomes identifies sex-specific resistome profiles. Gut Microbes. 2019;10(3):358–366. doi:10.1080/19490976.2018.1528822.
  • Santos-Marcos JA, Rangel-Zuniga OA, Jimenez-Lucena R, Quintana-Navarro GM, Garcia-Carpintero S, Malagon MM, Landa BB, Tena-Sempere M, Perez-Martinez P, Lopez-Miranda J, et al. Influence of gender and menopausal status on gut microbiota. Maturitas. 2018;116:43–53. doi:10.1016/j.maturitas.2018.07.008.
  • Mihajlovic J, Leutner M, Hausmann B, Kohl G, Schwarz J, Rover H, Stimakovits N, Wolf P, Maruszczak K, Bastian M, et al. Combined hormonal contraceptives are associated with minor changes in composition and diversity in gut microbiota of healthy women. Environ Microbiol. 2021;23(6):3037–3047. doi:10.1111/1462-2920.15517.
  • Thackray VS. Microbes, and polycystic ovary syndrome. Trends Endocrinol Metab. 2019;30(1):54–65. doi:10.1016/j.tem.2018.11.001.
  • Markle JG, Frank DN, Mortin-Toth S, Robertson CE, Feazel LM, Rolle-Kampczyk U, von Bergen M, McCoy KD, Macpherson AJ, Danska JS. Sex differences in the gut microbiome drive hormone-dependent regulation of autoimmunity. Science. 2013;339(6123):1084–1088. doi:10.1126/science.1233521.
  • Pernigoni N, Zagato E, Calcinotto A, Troiani M, Mestre RP, Cali B, Attanasio G, Troisi J, Minini M, Mosole S, et al. Commensal bacteria promote endocrine resistance in prostate cancer through androgen biosynthesis. Science. 2021;374(6564):216–224. doi:10.1126/science.abf8403.
  • Razavi AC, Potts KS, Kelly TN, Bazzano LA. Sex, gut microbiome, and cardiovascular disease risk. Biol Sex Differ. 2019;10(1):29. doi:10.1186/s13293-019-0240-z.
  • Audet MC. Stress-induced disturbances along the gut microbiota-immune-brain axis and implications for mental health: does sex matter? Front Neuroendocrinol. 2019;54:100772. doi:10.1016/j.yfrne.2019.100772.
  • Conforti F, Pala L, Bagnardi V, De Pas T, Martinetti M, Viale G, Gelber RD, Goldhirsch A. Cancer immunotherapy efficacy and patients’ sex: a systematic review and meta-analysis. Lancet Oncol. 2018;19(6):737–746. doi:10.1016/S1470-2045(18)30261-4.
  • Conforti F, Pala L, Bagnardi V, Viale G, De Pas T, Pagan E, Pennacchioli E, Cocorocchio E, Ferrucci PF, De Marinis F, et al. Sex-based heterogeneity in response to lung cancer immunotherapy: a systematic review and meta-analysis. J Natl Cancer Inst. 2019;111(8):772–781. doi:10.1093/jnci/djz094.
  • Yang C, Jin J, Yang Y, Sun H, Wu L, Shen M, Hong X, Li W, Lu L, Cao D, et al. Androgen receptor-mediated CD8(+) T cell stemness programs drive sex differences in antitumor immunity. Immunity. 2022;55(7):1268–83 e9. doi:10.1016/j.immuni.2022.05.012.
  • Guan X, Polesso F, Wang C, Sehrawat A, Hawkins RM, Murray SE, Thomas GV, Caruso B, Thompson RF, Wood MA, et al. Androgen receptor activity in T cells limits checkpoint blockade efficacy. Nature. 2022;606(7915):791–796. doi:10.1038/s41586-022-04522-6.
  • Yuan B, Clark CA, Wu B, Yang J, Drerup JM, Li T, Jin VX, Hu Y, Curiel TJ, Li R. Estrogen receptor beta signaling in CD8 + T cells boosts T cell receptor activation and antitumor immunity through a phosphotyrosine switch. J ImmunoTher Cancer. 2021;9(1):e001932. doi:10.1136/jitc-2020-001932.
  • Chakraborty B, Byemerwa J, Shepherd J, Haines CN, Baldi R, Gong W, Liu W, Mukherjee D, Artham S, Lim F, et al. Inhibition of estrogen signaling in myeloid cells increases tumor immunity in melanoma. J Clin Invest. 2021;131:131. doi:10.1172/JCI151347.
  • Paulos CM, Wrzesinski C, Kaiser A, Hinrichs CS, Chieppa M, Cassard L, Palmer DC, Boni A, Muranski P, Yu Z, et al. Microbial translocation augments the function of adoptively transferred self/tumor-specific CD8+ T cells via TLR4 signaling. J Clin Invest. 2007;117(8):2197–2204. doi:10.1172/JCI32205.
  • Viaud S, Saccheri F, Mignot G, Yamazaki T, Daillere R, Hannani D, Enot DP, Pfirschke C, Engblom C, Pittet MJ, et al. The intestinal microbiota modulates the anticancer immune effects of cyclophosphamide. Science. 2013;342(6161):971–976. doi:10.1126/science.1240537.
  • Iida N, Dzutsev A, Stewart CA, Smith L, Bouladoux N, Weingarten RA, Molina DA, Salcedo R, Back T, Cramer S, et al. Commensal bacteria control cancer response to therapy by modulating the tumor microenvironment. Science. 2013;342(6161):967–970. doi:10.1126/science.1240527.
  • Sivan A, Corrales L, Hubert N, Williams JB, Aquino-Michaels K, Earley ZM, Benyamin FW, Man Lei Y, Jabri B, Alegre M-L, et al. Commensal bifidobacterium promotes antitumor immunity and facilitates anti–pd-L1 efficacy. Science. 2015;350(6264):1084–1089. doi:10.1126/science.aac4255.
  • Vetizou M, Pitt JM, Daillere R, Lepage P, Waldschmitt N, Flament C, Rusakiewicz S, Routy B, Roberti MP, Duong CPM, et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science. 2015;350(6264):1079–1084. doi:10.1126/science.aad1329.
  • Gharaibeh RZ, Jobin C. Microbiota and cancer immunotherapy: in search of microbial signals. Gut. 2019;68(3):385–388. doi:10.1136/gutjnl-2018-317220.
  • Davar D, Dzutsev AK, McCulloch JA, Rodrigues RR, Chauvin JM, Morrison RM, Deblasio RN, Menna C, Ding Q, Pagliano O, et al. Fecal microbiota transplant overcomes resistance to anti–pd-1 therapy in melanoma patients. Science. 2021;371(6529):595–602. doi:10.1126/science.abf3363.
  • Baruch EN, Youngster I, Ben-Betzalel G, Ortenberg R, Lahat A, Katz L, Adler K, Dick-Necula D, Raskin S, Bloch N, et al. Fecal microbiota transplant promotes response in immunotherapy-refractory melanoma patients. Science. 2021;371(6529):602–609. doi:10.1126/science.abb5920.
  • Lu Y, Yuan X, Wang M, He Z, Li H, Wang J, Li Q. Gut microbiota influence immunotherapy responses: mechanisms and therapeutic strategies. J Hematol Oncol. 2022;15(1):47. doi:10.1186/s13045-022-01273-9.
  • Wu J, Wang S, Zheng B, Qiu X, Wang H, Chen L. Modulation of gut microbiota to enhance effect of checkpoint inhibitor immunotherapy. Front Immunol. 2021;12:669150. doi:10.3389/fimmu.2021.669150.
  • Griffin ME, Espinosa J, Becker JL, Luo JD, Carroll TS, Jha JK, Fanger GR, Hang HC. Enterococcus peptidoglycan remodeling promotes checkpoint inhibitor cancer immunotherapy. Science. 2021;373(6558):1040–1046. doi:10.1126/science.abc9113.
  • Kawanabe-Matsuda H, Takeda K, Nakamura M, Makino S, Karasaki T, Kakimi K, Nishimukai M, Ohno T, Omi J, Kano K, et al. Dietary lactobacillus-derived exopolysaccharide enhances immune-checkpoint blockade therapy. Cancer Discov. 2022;12(5):1336–1355. doi:10.1158/2159-8290.CD-21-0929.
  • Zhang W, Xu L, Park HB, Hwang J, Kwak M, Lee PCW, Liang G, Zhang X, Xu J, Jin J-O. Escherichia coli adhesion portion FimH functions as an adjuvant for cancer immunotherapy. Nat Commun. 2020;11(1):1187. doi:10.1038/s41467-020-15030-4.
  • Shi Y, Zheng W, Yang K, Harris KG, Ni K, Xue L, Lin W, Chang EB, Weichselbaum RR, Fu Y-X. Intratumoral accumulation of gut microbiota facilitates CD47-based immunotherapy via STING signaling. J Exp Med. 2020; 217(5). doi:10.1084/jem.20192282.
  • Ragone C, Manolio C, Mauriello A, Cavalluzzo B, Buonaguro FM, Tornesello ML, Tagliamonte M, Buonaguro L. Molecular mimicry between tumor associated antigens and microbiota-derived epitopes. J Transl Med. 2022;20(1):316. doi:10.1186/s12967-022-03512-6.
  • Nomura M, Nagatomo R, Inoue K, Doi K, Shimizu J, Baba K, Saito T, Matsumoto S, Muto M. Association of SCFA in gut microbiome and clinical response in solid cancer patients treated with andi-PD-1 antibody. Annals of Oncology. 2019;30:v509. doi:10.1093/annonc/mdz253.074.
  • Nomura M, Nagatomo R, Doi K, Shimizu J, Baba K, Saito T, Matsumoto S, Inoue K, Muto M. Association of short-chain fatty acids in the gut microbiome with clinical response to treatment with nivolumab or pembrolizumab in patients with solid cancer tumors. JAMA Netw Open. 2020;3(4):e202895. doi:10.1001/jamanetworkopen.2020.2895.
  • Botticelli A, Vernocchi P, Marini F, Quagliariello A, Cerbelli B, Reddel S, Del Chierico F, Di Pietro F, Giusti R, Tomassini A, et al. Gut metabolomics profiling of non-small cell lung cancer (NSCLC) patients under immunotherapy treatment. J Transl Med. 2020;18(1):49. doi:10.1186/s12967-020-02231-0.
  • Zheng Y, Wang T, Tu X, Huang Y, Zhang H, Tan D, Jiang W, Cai S, Zhao P, Song R, et al. Gut microbiome affects the response to anti-PD-1 immunotherapy in patients with hepatocellular carcinoma. J ImmunoTher Cancer. 2019;7(1):193. doi:10.1186/s40425-019-0650-9.
  • Peng Z, Cheng S, Kou Y, Wang Z, Jin R, Hu H, Zhang X, Gong J-F, Li J, Lu M, et al. The gut microbiome is associated with clinical response to anti–PD-1/PD-L1 immunotherapy in gastrointestinal cancer. Cancer Immunol Res. 2020;8(10):1251–1261. doi:10.1158/2326-6066.CIR-19-1014.
  • He Y, Fu L, Li Y, Wang W, Gong M, Zhang J, Dong X, Huang J, Wang Q, Mackay CR, et al. Gut microbial metabolites facilitate anticancer therapy efficacy by modulating cytotoxic CD8+ T cell immunity. Cell Metab. 2021;33(5):988–1000 e7. doi:10.1016/j.cmet.2021.03.002.
  • Mager LF, Burkhard R, Pett N, Cooke NCA, Brown K, Ramay H, Paik S, Stagg J, Groves RA, Gallo M, et al. Microbiome-derived inosine modulates response to checkpoint inhibitor immunotherapy. Science. 2020;369(6510):1481–1489. doi:10.1126/science.abc3421.
  • Wang T, Gnanaprakasam JNR, Chen X, Kang S, Xu X, Sun H, Liu L, Rodgers H, Miller E, Cassel TA, et al. Inosine is an alternative carbon source for CD8(+)-T-cell function under glucose restriction. Nat Metab. 2020;2(7):635–647. doi:10.1038/s42255-020-0219-4.
  • Wang H, Rong X, Zhao G, Zhou Y, Xiao Y, Ma D, Jin X, Wu Y, Yan Y, Yang H, et al. The microbial metabolite trimethylamine N-oxide promotes antitumor immunity in triple-negative breast cancer. Cell Metab. 2022;34(4):581–94 e8. doi:10.1016/j.cmet.2022.02.010.
  • Frankel AE, Coughlin LA, Kim J, Froehlich TW, Xie Y, Frenkel EP, Koh AY. Metagenomic shotgun sequencing and unbiased metabolomic profiling identify specific human gut microbiota and metabolites associated with immune checkpoint therapy efficacy in melanoma patients. Neoplasia. 2017;19(10):848–855. doi:10.1016/j.neo.2017.08.004.
  • Zhao H, Wang D, Zhang Z, Xian J, Bai X. Effect of gut microbiota-derived metabolites on immune checkpoint inhibitor therapy: enemy or friend? Molecules. 2022;27(15):4799. doi:10.3390/molecules27154799.
  • Jalandra R, Dalal N, Yadav AK, Verma D, Sharma M, Singh R, Khosla A, Kumar A, Solanki PR. Emerging role of trimethylamine-N-oxide (TMAO) in colorectal cancer. Appl Microbiol Biotechnol. 2021;105(20):7651–7660. doi:10.1007/s00253-021-11582-7.
  • Ajouz H, Mukherji D, Shamseddine A. Secondary bile acids: an underrecognized cause of colon cancer. World J Surg Oncol. 2014;12(1):164. doi:10.1186/1477-7819-12-164.
  • Xie G, Wang X, Zhao A, Yan J, Chen W, Jiang R, Ji J, Huang F, Zhang Y, Lei S, et al. Sex-dependent effects on gut microbiota regulate hepatic carcinogenic outcomes. Sci Rep. 2017;7(1):45232. doi:10.1038/srep45232.
  • Thomas MS, Fernandez ML. Trimethylamine N-Oxide (TMAO), diet and cardiovascular disease. Curr Atheroscler Rep. 2021;23(4):12. doi:10.1007/s11883-021-00910-x.
  • Botticelli A, Cerbelli B, Lionetto L, Zizzari I, Salati M, Pisano A, Federica M, Simmaco M, Nuti M, Marchetti P. Can IDO activity predict primary resistance to anti-PD-1 treatment in NSCLC? J Transl Med. 2018;16(1):219. doi:10.1186/s12967-018-1595-3.
  • Coutzac C, Jouniaux JM, Paci A, Schmidt J, Mallardo D, Seck A, Asvatourian V, Cassard L, Saulnier P, Lacroix L, et al. Systemic short chain fatty acids limit antitumor effect of CTLA-4 blockade in hosts with cancer. Nat Commun. 2020;11(1):2168. doi:10.1038/s41467-020-16079-x.
  • Wang L, Jiang G, Jing N, Liu X, Zhuang H, Zeng W, Liang W, Liu Z. Downregulating testosterone levels enhance immunotherapy efficiency. Oncoimmunology. 2021;10(1):1981570. doi:10.1080/2162402X.2021.1981570.
  • Pinato DJ, Gramenitskaya D, Altmann DM, Boyton RJ, Mullish BH, Marchesi JR, Bower M. Antibiotic therapy and outcome from immune-checkpoint inhibitors. J ImmunoTher Cancer. 2019;7(1):287. doi:10.1186/s40425-019-0775-x.
  • Villanueva L, Alvarez-Errico D, Esteller M. The contribution of epigenetics to cancer immunotherapy. Trends Immunol. 2020;41(8):676–691. doi:10.1016/j.it.2020.06.002.
  • Davar D, Zarour HM. Facts and hopes for gut microbiota interventions in cancer immunotherapy. Clin Cancer Res. 2022;28(20):4370–4384. doi:10.1158/1078-0432.CCR-21-1129.
  • Rubin JB, Lagas JS, Broestl L, Sponagel J, Rockwell N, Rhee G, Rosen SF, Chen S, Klein RS, Imoukhuede P, et al. Sex differences in cancer mechanisms. Biol Sex Differ. 2020;11(1):17. doi:10.1186/s13293-020-00291-x.
  • Klein SL, Jedlicka A, Pekosz A. The Xs and Y of immune responses to viral vaccines. Lancet Infect Dis. 2010;10(5):338–349. doi:10.1016/S1473-3099(10)70049-9.
  • Klein SL, Flanagan KL. Sex differences in immune responses. Nat Rev Immunol. 2016;16(10):626–638. doi:10.1038/nri.2016.90.
  • Wu Y, Ju Q, Jia K, Yu J, Shi H, Wu H, Jiang M. Correlation between sex and efficacy of immune checkpoint inhibitors (PD-1 and CTLA-4 inhibitors). Int J Cancer. 2018;143(1):45–51. doi:10.1002/ijc.31301.
  • Wang C, Qiao W, Jiang Y, Zhu M, Shao J, Ren P, Liu D, Li W. Effect of sex on the efficacy of patients receiving immune checkpoint inhibitors in advanced non-small cell lung cancer. Cancer Med. 2019;8(8):4023–4031. doi:10.1002/cam4.2280.
  • Pinto JA, Vallejos CS, Raez LE, Mas LA, Ruiz R, Torres-Roman JS, Morante Z, Araujo JM, Gómez HL, Aguilar A, et al. Gender and outcomes in non-small cell lung cancer: an old prognostic variable comes back for targeted therapy and immunotherapy? ESMO Open. 2018;3(3):e000344. doi:10.1136/esmoopen-2018-000344.
  • Litchfield K, Reading JL, Puttick C, Thakkar K, Abbosh C, Bentham R, Watkins TBK, Rosenthal R, Biswas D, Rowan A, et al. Meta-analysis of tumor- and T cell-intrinsic mechanisms of sensitization to checkpoint inhibition. Cell. 2021;184(3):596–614 e14. doi:10.1016/j.cell.2021.01.002.
  • Ye Y, Jing Y, Li L, Mills GB, Diao L, Liu H, Han L. Sex-associated molecular differences for cancer immunotherapy. Nat Commun. 2020;11(1):1779. doi:10.1038/s41467-020-15679-x.
  • Wallis CJD, Butaney M, Satkunasivam R, Freedland SJ, Patel SP, Hamid O, Pal SK, Klaassen Z. Association of patient sex with efficacy of immune checkpoint inhibitors and overall survival in advanced cancers: a systematic review and meta-analysis. JAMA Oncol. 2019;5(4):529–536. doi:10.1001/jamaoncol.2018.5904.
  • Sinha N, Sinha S, Cheng K, Madan S, Erez A, Ryan BM, Schäffer AA, Aldape K, Ruppin E. Using a recently approved tumor mutational burden biomarker to stratify patients for immunotherapy may introduce a sex bias. Jco Precis Oncol. 2021;5(5):1147–1150. doi:10.1200/PO.21.00168.
  • Pu Y, Xu M, Liang Y, Yang K, Guo Y, Yang X, Fu Y-X. Androgen receptor antagonists compromise T cell response against prostate cancer leading to early tumor relapse. Sci Transl Med. 2016;8(333):333ra47. doi:10.1126/scitranslmed.aad5659.
  • Haupt S, Caramia F, Klein SL, Rubin JB, Haupt Y. Sex disparities matter in cancer development and therapy. Nat Rev Cancer. 2021;21(6):393–407. doi:10.1038/s41568-021-00348-y.
  • Ozdemir BC, Dotto G-P. Sex hormones and anticancer immunity. Clin Cancer Res. 2019;25(15):4603–4610. doi:10.1158/1078-0432.CCR-19-0137.
  • Tulchiner G, Pichler R, Ulmer H, Staudacher N, Lindner AK, Brunner A, Zelger B, Steinkohl F, Aigner F, Horninger W, et al. Sex-specific hormone changes during immunotherapy and its influence on survival in metastatic renal cell carcinoma. Cancer Immunol Immunother. 2021;70(10):2805–2817. doi:10.1007/s00262-021-02882-y.
  • Valeri F, Endres K. How biological sex of the host shapes its gut microbiota. Front Neuroendocrinol. 2021;61:100912. doi:10.1016/j.yfrne.2021.100912.
  • Fransen F, van Beek AA, Borghuis T, Meijer B, Hugenholtz F, van der Gaast-de Jongh C, Savelkoul HF, de Jonge MI, Faas MM, Boekschoten MV, et al. The impact of gut microbiota on gender-specific differences in immunity. Front Immunol. 2017;8:754. doi:10.3389/fimmu.2017.00754.
  • Org E, Mehrabian M, Parks BW, Shipkova P, Liu X, Drake TA, Lusis AJ. Sex differences and hormonal effects on gut microbiota composition in mice. Gut Microbes. 2016;7(4):313–322. doi:10.1080/19490976.2016.1203502.
  • Kozik AJ, Nakatsu CH, Chun H, Jones-Hall YL. Age, sex, and TNF associated differences in the gut microbiota of mice and their impact on acute TNBS colitis. Exp Mol Pathol. 2017;103(3):311–319. doi:10.1016/j.yexmp.2017.11.014.
  • Takagi T, Naito Y, Inoue R, Kashiwagi S, Uchiyama K, Mizushima K, Tsuchiya S, Dohi O, Yoshida N, Kamada K, et al. Differences in gut microbiota associated with age, sex, and stool consistency in healthy Japanese subjects. J Gastroenterol. 2019;54(1):53–63. doi:10.1007/s00535-018-1488-5.
  • Gao X, Zhang M, Xue J, Huang J, Zhuang R, Zhou X, Zhang H, Fu Q, Hao Y. Body mass index differences in the gut microbiota are gender specific. Front Microbiol. 2018;9:1250. doi:10.3389/fmicb.2018.01250.
  • Borgo F, Garbossa S, Riva A, Severgnini M, Luigiano C, Benetti A, Pontiroli AE, Morace G, Borghi E. Body mass index and sex affect diverse microbial niches within the gut. Front Microbiol. 2018;9:213. doi:10.3389/fmicb.2018.00213.
  • Yang T, Santisteban MM, Rodriguez V, Li E, Ahmari N, Carvajal JM, Zadeh M, Gong M, Qi Y, Zubcevic J, et al. Gut dysbiosis is linked to hypertension. Hypertension. 2015;65(6):1331–1340. doi:10.1161/HYPERTENSIONAHA.115.05315.
  • Magne F, Gotteland M, Gauthier L, Zazueta A, Pesoa S, Navarrete P, Balamurugan R. The firmicutes/bacteroidetes ratio: a relevant marker of gut dysbiosis in obese patients? Nutrients. 2020;12(5):1474. doi:10.3390/nu12051474.
  • Koliada A, Moseiko V, Romanenko M, Lushchak O, Kryzhanovska N, Guryanov V, Vaiserman A. Sex differences in the phylum-level human gut microbiota composition. BMC Microbiol. 2021;21(1):131. doi:10.1186/s12866-021-02198-y.
  • Dominianni C, Sinha R, Goedert JJ, Pei Z, Yang L, Hayes RB, Ahn J. Sex, body mass index, and dietary fiber intake influence the human gut microbiome. PloS One. 2015;10(4):e0124599. doi:10.1371/journal.pone.0124599.
  • Falony G, Joossens M, Vieira-Silva S, Wang J, Darzi Y, Faust K, Kurilshikov A, Bonder MJ, Valles-Colomer M, Vandeputte D, et al. Population-level analysis of gut microbiome variation. Science. 2016;352(6285):560–564. doi:10.1126/science.aad3503.
  • Manor O, Dai CL, Kornilov SA, Smith B, Price ND, Lovejoy JC, Gibbons SM, Magis AT. Health and disease markers correlate with gut microbiome composition across thousands of people. Nat Commun. 2020;11(1):5206. doi:10.1038/s41467-020-18871-1.
  • Suzuki TA, Worobey M. Geographical variation of human gut microbial composition. Biol Lett. 2014;10(2):20131037. doi:10.1098/rsbl.2013.1037.
  • Shin JH, Park YH, Sim M, Kim SA, Joung H, Shin DM. Serum level of sex steroid hormone is associated with diversity and profiles of human gut microbiome. Res Microbiol. 2019;170(4–5):192–201. doi:10.1016/j.resmic.2019.03.003.
  • Liao H, Li C, Ai Y, Kou Y. Gut microbiome is more stable in males than in females during the development of colorectal cancer. J Appl Microbiol. 2021;131(1):435–448. doi:10.1111/jam.14943.
  • Mayneris-Perxachs J, Arnoriaga-Rodriguez M, Luque-Cordoba D, Priego-Capote F, Perez-Brocal V, Moya A, Burokas A, Maldonado R, Fernández-Real J-M. Gut microbiota steroid sexual dimorphism and its impact on gonadal steroids: influences of obesity and menopausal status. Microbiome. 2020;8(1):136. doi:10.1186/s40168-020-00913-x.
  • d’Afflitto M, Upadhyaya A, Green A, Peiris M. Association between sex hormone levels and gut microbiota composition and diversity—a systematic review. J Clin Gastroenterol. 2022;56(5):384–392. doi:10.1097/MCG.0000000000001676.
  • Flores R, Shi J, Fuhrman B, Xu X, Veenstra TD, Gail MH, Gajer P, Ravel J, Goedert JJ. Fecal microbial determinants of fecal and systemic estrogens and estrogen metabolites: a cross-sectional study. J Transl Med. 2012;10(1):253. doi:10.1186/1479-5876-10-253.
  • Fuhrman BJ, Feigelson HS, Flores R, Gail MH, Xu X, Ravel J, Goedert JJ. Associations of the fecal microbiome with urinary estrogens and estrogen metabolites in postmenopausal women. J Clin Endocrinol Metab. 2014;99(12):4632–4640. doi:10.1210/jc.2014-2222.
  • Sherman SB, Sarsour N, Salehi M, Schroering A, Mell B, Joe B, Hill JW. Prenatal androgen exposure causes hypertension and gut microbiota dysbiosis. Gut Microbes. 2018;9:1–22. doi:10.1080/19490976.2018.1441664.
  • Torres PJ, Skarra DV, Ho BS, Sau L, Anvar AR, Kelley ST, Thackray VG. Letrozole treatment of adult female mice results in a similar reproductive phenotype but distinct changes in metabolism and the gut microbiome compared to pubertal mice. BMC Microbiol. 2019;19(1):57. doi:10.1186/s12866-019-1425-7.
  • Menon R, Watson SE, Thomas LN, Allred CD, Dabney A, Azcarate-Peril MA, Sturino JM. Diet complexity and estrogen receptor β status affect the composition of the murine intestinal microbiota. Appl Environ Microbiol. 2013;79(18):5763–5773. doi:10.1128/AEM.01182-13.
  • Harada N, Hanaoka R, Hanada K, Izawa T, Inui H, Yamaji R. Hypogonadism alters cecal and fecal microbiota in male mice. Gut Microbes. 2016;7(6):533–539. doi:10.1080/19490976.2016.1239680.
  • Kaliannan K, Robertson RC, Murphy K, Stanton C, Kang C, Wang B, Hao L, Bhan AK, Kang JX. Estrogen-mediated gut microbiome alterations influence sexual dimorphism in metabolic syndrome in mice. Microbiome. 2018;6(1):205. doi:10.1186/s40168-018-0587-0.
  • Nuriel-Ohayon M, Neuman H, Ziv O, Belogolovski A, Barsheshet Y, Bloch N, Uzan A, Lahav R, Peretz A, Frishman S, et al. Progesterone increases bifidobacterium relative abundance during late pregnancy. Cell Rep. 2019;27(3):730–6 e3. doi:10.1016/j.celrep.2019.03.075.
  • Flak MB, Neves JF, Blumberg RI. Welcome to the microgenderome. Science. 2013;339(6123):1044–1045. doi:10.1126/science.1236226.
  • Yurkovetskiy L, Burrows M, Khan AA, Graham L, Volchkov P, Becker L, Antonopoulos D, Umesaki Y, Chervonsky A. Gender bias in autoimmunity is influenced by microbiota. Immunity. 2013;39(2):400–412. doi:10.1016/j.immuni.2013.08.013.
  • Collden H, Landin A, Wallenius V, Elebring E, Fandriks L, Nilsson ME, Ryberg H, Poutanen M, Sjögren K, Vandenput L, et al. The gut microbiota is a major regulator of androgen metabolism in intestinal contents. Am J Physiol Endocrinol Metab. 2019;317(6):E1182–92. doi:10.1152/ajpendo.00338.2019.
  • Baker JM, Al-Nakkash L, Herbst-Kralovetz MM. Estrogen-gut microbiome axis: physiological and clinical implications. Maturitas. 2017;103:45–53. doi:10.1016/j.maturitas.2017.06.025.
  • Pollet RM, D’agostino EH, Walton WG, Xu Y, Little MS, Biernat KA, Pellock SJ, Patterson LM, Creekmore BC, Isenberg HN, et al. An atlas of β-glucuronidases in the human intestinal microbiome. Structure. 2017;25(7):967–77 e5. doi:10.1016/j.str.2017.05.003.
  • Ervin SM, Li H, Lim L, Roberts LR, Liang X, Mani S, Redinbo MR. Gut microbial β-glucuronidases reactivate estrogens as components of the estrobolome that reactivate estrogens. J Biol Chem. 2019;294(49):18586–18599. doi:10.1074/jbc.RA119.010950.
  • Lombardi P, Goldin B, Boutin E, Gorbach SL. Metabolism of androgens and estrogens by human fecal microorganisms. J Steroid Biochem. 1978;9(8):795–801. doi:10.1016/0022-4731(78)90203-0.
  • Garcia-Gomez E, Gonzalez-Pedrajo B, Camacho-Arroyo I. Role of sex steroid hormones in bacterial-host interactions. Biomed Res Int. 2013;2013:928290. doi:10.1155/2013/928290.
  • Vich Vila A, Collij V, Sanna S, Sinha T, Imhann F, Bourgonje AR, Mujagic Z, Jonkers DMAE, Masclee AAM, Fu J, et al. Impact of commonly used drugs on the composition and metabolic function of the gut microbiota. Nat Commun. 2020;11(1):362. doi:10.1038/s41467-019-14177-z.
  • Ojanotko-Harri A, Nikkari T, Harri MP, Paunio KU. Metabolism of progesterone and testosterone by bacillus cereus strain socransky 67 and streptococcus mutans strain ingbritt. Oral Microbiol Immunol. 1990;5(4):237–239. doi:10.1111/j.1399-302X.1990.tb00653.x.
  • Everard A, Belzer C, Geurts L, Ouwerkerk JP, Druart C, Bindels LB, Guiot Y, Derrien M, Muccioli GG, Delzenne NM, et al. Cross-talk between akkermansia muciniphila and intestinal epithelium controls diet-induced obesity. Proc Natl Acad Sci U S A. 2013;110(22):9066–9071. doi:10.1073/pnas.1219451110.
  • Paone P, Cani PD. Mucus barrier, mucins and gut microbiota: the expected slimy partners? Gut. 2020;69(12):2232–2243. doi:10.1136/gutjnl-2020-322260.
  • Tremellen K. Gut Endotoxin Leading to a Decline in Gonadal function (GELDING) - a novel theory for the development of late onset hypogonadism in obese men. Basic Clin Androl. 2016;26(1):7. doi:10.1186/s12610-016-0034-7.
  • Tremellen K, McPhee N, Pearce K, Benson S, Schedlowski M, Engler H. Endotoxin-initiated inflammation reduces testosterone production in men of reproductive age. Am J Physiol Endocrinol Metab. 2018;314(3):E206–13. doi:10.1152/ajpendo.00279.2017.
  • Tian X, Yu Z, Feng P, Ye Z, Li R, Liu J, Hu J, Kakade A, Liu P, Li X. Lactobacillus plantarum TW1-1 alleviates diethylhexylphthalate-induced testicular damage in mice by modulating gut microbiota and decreasing inflammation. Front Cell Infect Microbiol. 2019;9:221. doi:10.3389/fcimb.2019.00221.
  • Zhang J, Sun Z, Jiang S, Bai X, Ma C, Peng Q, Chen K, Chang H, Fang T, Zhang H, et al. Probiotic bifidobacterium lactis v9 regulates the secretion of sex hormones in polycystic ovary syndrome patients through the gut-brain axis. mSystems. 2019;4(2). doi:10.1128/mSystems.00017-19.
  • Tong D. Selective estrogen receptor modulators contribute to prostate cancer treatment by regulating the tumor immune microenvironment. J ImmunoTher Cancer. 2022;10(4):e002944. doi:10.1136/jitc-2021-002944.
  • Ardiani A, Farsaci B, Rogers CJ, Protter A, Guo Z, King TH, Apelian D, Hodge JW. Combination therapy with a second-generation androgen receptor antagonist and a metastasis vaccine improves survival in a spontaneous prostate cancer model. Clin Cancer Res. 2013;19(22):6205–6218. doi:10.1158/1078-0432.CCR-13-1026.
  • Huang H, Zhou J, Chen H, Li J, Zhang C, Jiang X, Ni C. The immunomodulatory effects of endocrine therapy in breast cancer. J Exp Clin Cancer Res. 2021;40(1):19. doi:10.1186/s13046-020-01788-4.
  • Song CH, Kim N, Nam RH, Choi SI, Jang JY, Kim JW, Na HY, Lee H-N. Combination treatment with 17β-estradiol and anti-PD-L1 suppresses MC38 tumor growth by reducing PD-L1 expression and enhancing M1 macrophage population in MC38 colon tumor model. Cancer Lett. 2022;543:215780. doi:10.1016/j.canlet.2022.215780.
  • Gao G, Ma T, Zhang T, Jin H, Li Y, Kwok LY, Zhang H, Sun Z. Adjunctive probiotic lactobacillus rhamnosus probio-M9 administration enhances the effect of anti-PD-1 antitumor therapy via restoring antibiotic-disrupted gut microbiota. Front Immunol. 2021;12:772532. doi:10.3389/fimmu.2021.772532.
  • Takada K, Shimokawa M, Takamori S, Shimamatsu S, Hirai F, Tagawa T, Okamoto T, Hamatake M, Tsuchiya-kawano Y, Otsubo K, et al. Clinical impact of probiotics on the efficacy of anti-PD-1 monotherapy in patients with nonsmall cell lung cancer: a multicenter retrospective survival analysis study with inverse probability of treatment weighting. Int J Cancer. 2021;149(2):473–482. doi:10.1002/ijc.33557.
  • Spencer CN, McQuade JL, Gopalakrishnan V, McCulloch JA, Vetizou M, Cogdill AP, Khan MAW, Zhang X, White MG, Peterson CB, et al. Dietary fiber and probiotics influence the gut microbiome and melanoma immunotherapy response. Science. 2021;374(6575):1632–1640. doi:10.1126/science.aaz7015.
  • Gholizadeh Shamasbi S, Dehgan P, Mohammad-Alizadeh Charandabi S, Aliasgarzadeh A, Mirghafourvand M. The effect of resistant dextrin as a prebiotic on metabolic parameters and androgen level in women with polycystic ovarian syndrome: a randomized, triple-blind, controlled, clinical trial. Eur J Nutr. 2019;58(2):629–640. doi:10.1007/s00394-018-1648-7.