Next Article in Journal
Special Issue: “Management of Early Stage Cervical Cancer”
Previous Article in Journal
Surveillance of Low-Grade Non-Muscle Invasive Bladder Tumors Using Uromonitor: SOLUSION Trial
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Communication

Antibiotics Significantly Decrease the Survival of Head and Neck Carcinoma Patients with Immunotherapy: A Real-World Analysis of More Than 3000 Cases

1
Department of Oral and Maxillofacial Surgery, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
2
Institute of Physiology and Science-IT, Charité—Universitätsmedizin Berlin, Philippstr. 12, 10115 Berlin, Germany
3
Clinic for Otorhinolaryngology, Head and Neck Surgery, Klinikum Rechts der Isar der Technischen Universität München (MRI TUM), Ismaningerstr. 22, 81675 Munich, Germany
*
Author to whom correspondence should be addressed.
Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health.
Cancers 2023, 15(8), 2342; https://doi.org/10.3390/cancers15082342
Submission received: 20 March 2023 / Revised: 9 April 2023 / Accepted: 15 April 2023 / Published: 18 April 2023
(This article belongs to the Topic Gut Microbiota and Cancer)

Abstract

:

Simple Summary

It is well known that antibiotics alter the gut microbiome, and because it plays a role in drug metabolism, alterations to the microbiome may lead to ineffective immunotherapy in cancer patients. We investigated a real-world cohort of oral cancer patients who received immunotherapy. Patients were matched for age, sex, BMI, metastases, alcohol and nicotine dependence and sepsis to create two comparable groups. Patients who received antibiotics had a significantly decreased survival compared to those who did not. We believe that this finding is associated with less effective immunotherapy due to antibiotic-related changes in the gut microbiome.

Abstract

Objective: The human gut microbiome is strongly influenced by the administration of drugs, namely antibiotics. We hypothesized that the effectiveness of immunotherapy with pembrolizumab in oral squamous cell carcinoma patients is decreased by the administration of antibiotics three months before and after immunotherapy. Methods: We retrieved data from patients diagnosed with head and neck squamous cell carcinoma (HNSCC) (International Classification of Diseases [ICD]-10 codes C00-C14) and receiving immunotherapy with pembrolizumab from the TriNetX network. Two cohorts were built: patients in cohort I did not receive any antibiotics within three months before or up to three months after immunotherapy, while patients in cohort II were administered antibiotics at least once within three months before or after immunotherapy. To exclude confounders, we matched cohorts 1:1 for age, sex, secondary lymph node metastases, nicotine dependence, the insertion of feeding devices, body mass index (BMI) and severe sepsis. After defining the primary outcome as “death”, a Kaplan–Meier analysis was performed, and the risk ratio (RR), odds ratio (OR) and hazard ratio (HR) were calculated. Results: A total of 3651 patients were enrolled, and after matching, each cohort consisted of 1362 patients. Among cohorts I and II, 346 and 511 patients were deceased within one year (risk of death = 25.5 and 38.3%, respectively), whereby the risk difference was significant (p = 0.000; log-rank test). The RR was 0.68 (95% confidence interval: 0.60–0.76), OR was 0.57 (0.48–0.67) and HR was 0.58 (0.51–0.67). Conclusions: Our hypothesis was confirmed: administering antibiotics significantly decreases the drug effectiveness of immunotherapy. We hypothesize that this finding is associated with antibiotic-related changes in the gut microbiome. Prospective clinical studies on the gut microbiome in cancer patients are necessary to understand the complex ecosystem of microbiota during immunotherapy. Trial Registration: Due to the retrospective nature of the study, no registration was necessary.

1. Introduction

Head and neck squamous cell carcinoma (HNSCC), among the most common cancers [1], is mainly caused by risk factors such as smoking and alcohol consumption. Despite intensive efforts, this cancer not been significantly reduced in recent decades. The current primary treatment regimen consists of surgery or radiation with or without chemotherapy [2]. In the setting of first-line treatment failure, the administration of PD-1 to target immune checkpoint inhibitors (ICIs), such as pembrolizumab (with chemotherapy or as a monotherapy) [3,4] or nivolumab [5], is well established and has become an integral part of treatment guidelines [6,7]. The benefit of these new agents is a reduction in severe treatment side effects, commonly associated with an extreme chemotherapy/Erbitux regimen, which leads to a massively improved quality of life, as well as significantly increased survival rates. Depending on the combined positive score (CPS) that reflects PD-L1 expression on tumor cells, infiltrating lymphocytes and macrophages, overall survival (OS) was significantly prolonged in patients receiving pembrolizumab compared to those receiving cetuximab–chemotherapy: PD-L1 CPS ≥ 20 and CPS ≥ 1, respectively. Pembrolizumab alone and pembrolizumab–chemotherapy also demonstrated a substantially longer duration of response (DOR) in all populations [8]. Similar effects can be observed in real-world treatment data on nivolumab monotherapy [9]. When comparing all ICI response rates in squamous cell cancers, it is striking that recurrent/metastatic head and neck cancers (R/M HNSCCs) are found within the lowest third of all cancers and have a response rate of 15–20%. To date, there is no obvious molecular or immunological reason why this response rate to ICI in R/M HNSCC is so low [10].
One possible explanation could be the timepoint of ICI treatment, which, to date, has only been available for patients in first-line failure settings such as recurrences or metastases. It is well-recognized that recurrent/metastatic cancers do not share clonal similarities with primary cancers in HNSCC [11,12]. New neoadjuvant treatment studies are already revealing different response rates to ICI [13,14,15].
Another reason could be the specifically altered microbiome of HNSCC patients. Very early on in the checkpoint treatment, several groups published data which found that primary resistance to ICIs can be attributed to abnormal gut microbiome composition, thereby [16,17] unravelling the importance of the commensal microbiota in immuno-oncology. Meanwhile, it is well known that this complex ecosystem is influenced by various drugs and can also influence treatment response [18,19]. In several pre-clinical models, the absence of an intact gut microbiome adversely affected ICI efficacy. So far in HNSCC, it remains unclear whether antibiotic-induced dysbiosis influences the clinical response through the modulation of the gut microbiome or whether it constitutes an additional surrogate marker of unfit or immunodeficient patients [20,21].
To investigate the influence of antibiotics on the clinical outcome of pembrolizumab treatment in a large real-world cohort, we selected the TriNetX Global Health Research Network (TriNetX, Cambridge, MA, USA) to take a closer look at patients’ data. The TriNetX provides access to a significant number of medical records from more than 78 healthcare organizations (HCOs) in 11 countries. Its intent is to bring together HCOs, contract research institutes and pharmaceutical companies to collect and exchange longitudinal clinical data and provide state-of-the-art statistical analytics. As of April 2023, TriNetX had collated electronic medical records of more than 250 million individuals. The network had previously been used to research medical topics of global importance, including the COVID-19 pandemic [22]. Detailed data analyses were performed to improve understanding of how the use of antibiotics before or after ICI therapy might change clinical prognoses.

2. Patients and Methods

2.1. Data Acquisition, Allocation and Matching

We retrieved data from patients diagnosed with head and neck squamous cell carcinoma (HNSCC) (International Classification of Diseases [ICD]-10 codes C00-C14) and receiving immunotherapy with pembrolizumab from the TriNetX network. Two cohorts were built: patients in cohort I did not receive any antibiotics within three months before or up to three months after immunotherapy, while patients in cohort II were administered antibiotics at least once within three months before or after immunotherapy. To exclude confounders, we matched cohorts 1:1 for age, sex, secondary lymph node metastases, nicotine dependence, insertion of feeding devices and body mass index (BMI).

2.2. Data Analysis

After defining the primary outcome as “death”, the time window was set to one year after the index event (ICD-10 codes C00-C14). Outcome events were recorded on a daily interval. Subsequently, a Kaplan–Meier analysis was performed, and the risk ratio (RR), odds ratio (OR) and hazard ratio (HR) were calculated. Statistical analysis was performed using the log-rank test, where p ≤ 0.05 was defined as significant. For 1:1 matching, a propensity score-matching algorithm was used. The system generated propensity scores for each patient in each cohort using logistic regression (software package scikit-learn). The propensity score ranged between 0 and 1 and indicated the predicted probability that a patient was in cohort I or II given the patient’s covariates. The greedy nearest-neighbor matching algorithm with a caliper of 0.1 pooled standard deviation was used. The caliper of 0.1 means that patients with very different propensity scores are not matched.
Furthermore, treatment pathways for the antibiotics used in cohort II were investigated.

3. Results

3.1. Assessment, Allocation and Matching

The access date was 6 April 2023, so no patients had to be excluded for index events that were more than 20 years old. A total of 42 HCOs responded with patients, and 3651 patients were enrolled. After matching for age, sex, secondary lymph node metastases, nicotine dependence, insertion of feeding devices, body mass index (BMI) and severe sepsis, each cohort accounted for 1362 patients. After matching, the cohorts did not differ significantly from each other (p > 0.05), as illustrated in Table 1.
Details about the assessment, allocation and matching can be found in the modified Consort diagram (Figure 1), as well as in Table 2.

3.2. Survival Analysis

In cohorts I and II, 346 and 511 patients, respectively, died within one year (risk of death = 25.4 and 37.5%), for which the risk difference was significant (p < 0.0001; log-rank test). The RR was 0.68 (95% confidence interval (CI) 0.60–0.76), OR was 0.57 (CI, 0.48–0.67) and HR was 0.58 (0.51–0.67).
Table 3 includes patients in each cohort with the outcome (death in the cohort and the number of patients that had the outcome in the time window), median survival (the number of days when the survival dropped below 50%; “--” indicates that survival did not drop below 50% during the time window of one year) and survival probability at the end of the time window (% survival at the end of the time window). In addition, a log-rank test, hazard ratio and z-test for proportionality were performed.

3.3. Treatment Pathways

For cohort II, a treatment pathway analysis was run. In the first line of treatment (LOT 1), the most administered regimen was erythromycin, followed by a combination therapy of extended spectrum penicillins and cephalosporins (third generation). Quinolones are more present in the second line of treatment (Figure 2).

4. Discussion

In concordance with many publications in recent years, our data confirmed that there is a crucial balance between human health and disease mediated by the gut microbiome, which modulates the host immune system both locally and systemically [23,24].
Especially in the context of immunotherapy, the gut microbiome appears to be among the most dominant biological markers for distinguishing therapy-responding patients from non-responders in various types of immunotherapies [25,26,27,28,29,30].
Although not understood in mechanistic details, emerging evidence indicates that disruption of the intestinal flora by different antibiotics can result in several negative consequences for the gut microbiota: the reduced diversity of species and an alteration in metabolic activity and the selection of antibiotic-resistant organisms [31,32]. This fact is not only important during the early assembly of the gut microbiome during neonatal growth [33] but especially during cancer therapy, such as surgery in non-sterile areas such as the head and neck region or during radiotherapy [34,35] and in immunotherapy especially [36]. The sample size of those studies reviewed in meta-analyses is usually low, which motivated the authors of our study to analyze a large database of malignant neoplasms of the head and neck. In total, 1502 datasets were available for patients with antibiotics, and 2149 datasets were available for those without antibiotics during or before immunotherapy. By exploiting the TriNetX Global Health Research Network, we can prove that the drug effectiveness of immunotherapy is decreased by the administration of antibiotics. Individuals who were administered antibiotics within ±3 months of immunotherapy had a significantly higher risk of dying within one year compared to individuals who were not administered antibiotics.
There are studies analyzing the degree of perturbation of the commensal microbiome by four commonly used antibiotics: azithromycin, levofloxacin, cefpodoxime and combinations thereof [37]. Consequently, if antibiotic therapy cannot be avoided in immunotherapy, certain drug regimens such as azithromycin and its combinations should be avoided as they have been shown to delay for two months the recovery of species richness. Whether probiotic administration or an additional immunotherapy delay improves the outcome of anti-cancer immunotherapy needs to be examined in prospective clinical trials. Furthermore, it is unclear whether the parameters used to characterize the gut microbiome, such as species richness or aerobic/anaerobic composition, are relevant to the differences in the observed outcomes. Mechanistic studies could help identify certain relevant species, (biosynthetic) pathways, messengers, metabolites or other supportive effects of the microbiome.
Alongside the applied methods are specific limitations regarding the results that need to be addressed. Most importantly, we had no data on the microbiome of the patients, so we used the administration of antibiotics as a marker for an altered gut microbiome [38]. By matching the cohorts for age, sex, lymph node metastases, BMI, feeding device, smoking habits and severe sepsis, we designed two cohorts that were as similar as possible. A disease-specific survival analysis would be interesting, but due to the de-identified data, we had no data on individual causes of death. It must be mentioned that tobacco use is self-reported, and we had no detailed data on tobacco use (e.g., pack-years) [39]. A certain risk of confounders, such as metabolic alterations after alcohol consumption, an important lifestyle factor frequently associated with smoking, has yet to be comprehensively investigated, so a certain bias should be considered [40]. Another important limitation was the lack of information on the form of antibiotic administration: oral or intravenous. Depending on how the drug was metabolized, agents with no or low bile or fecal excretion were available and may have affected the gut microbiome less. The implications of drug excretion on the gut microbiome are still controversial [41].
Future studies may apply a prospective approach to include data, especially on the microbiome before, during and after treatment, and smoking/drinking behavior. Furthermore, tumor status (e.g., HPV+) should include lifestyle factors and the form of drug administration. Particularly in HNSCC reconstructive surgery, long phases of parental tube feeding might add an additional load to gut dysbiosis, so special care should be applied with regard to the length and composition of the applied nutrients.

5. Conclusions

In summary, our data impressively underlined the assumptions of studies connecting antibiotics-induced dysbiosis and therapeutic outcomes. At the very least, the harvested data support our hypotheses of decreased therapy effectiveness due to antibiotic use. This might encourage further research on the gut and locoregional microbiome in head and neck cancer patients. If the results could be confirmed in the future, the microbiome should be considered in individualized cancer therapy, e.g., for applying supportive probiotics when antibiotics must be administered [42].

Author Contributions

Conceptualization: S.P., M.H. and B.W.; methodology: S.P. and R.P.; software: S.P. and R.P.; validation: S.P., M.H., B.W. and M.W.; data curation: S.P., R.P., B.W. and M.W.; writing—original draft: S.P.; writing—review and editing: S.P., R.P., M.H., B.W. and M.W.; visualization: S.P.; project administration: S.P. and R.P.; supervision and funding acquisition: R.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Deutsche Forschungsgemeinschaft (grant number TRR295).

Institutional Review Board Statement

All methods were carried out in accordance with relevant guidelines and regulations. All HCOs from which data were reported to TriNetX obtained informed consent in written form from all patients or their legal guardians. TriNetX is compliant with the Health Insurance Portability and Accountability Act (HIPAA), the U.S. federal law that protects the privacy and security of healthcare data. TriNetX is certified to the ISO 27001:2013 standard and maintains an Information Security Management System (ISMS) to ensure the protection of the healthcare data it has access to and to meet the requirements of the HIPAA Security Rule. Any aggregate data displayed on the TriNetX Platform, or any patient-level data in a dataset generated by the TriNetX Platform, contains only de-identified data per the de-identification standard defined in Section §164.514(a) of the HIPAA Privacy Rule. The process by which the data is de-identified is attested to through a formal determination by a qualified expert as defined in Section §164.514(b)(1) of the HIPAA Privacy Rule. This formal determination by a qualified expert, updated in December 2020, supersedes the need for TriNetX’s previous waiver from the Western Institutional Review Board (IRB). The TriNetX network contains data provided by participating Healthcare Organizations (HCOs), each of which represent and warrant that it has all necessary rights, consents, approvals and authority to provide data to TriNetX under a Business Associate Agreement (BAA), so long as their name remains anonymous as a data source and their data are utilized for research purposes. The data shared through the TriNetX Platform are attenuated to ensure that they do not include sufficient information to facilitate the determination of which HCO contributed which specific piece of information about a patient (https://trinetx.com/trinetx-publication-guidelines/, accessed on 5 May 2022). Since data were exclusively retrieved from TriNetX, an exception was granted by the institutional ethics committee, and no approval was necessary.

Data Availability Statement

The datasets used and analyzed can be retrieved from the TriNetX network (https://trinetx.com, accessed on 5 May 2022). If no access is available, the datasets can be retrieved from the corresponding author based on reasonable request.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

HCOheath care organization
HPVhuman papillomavirus
ICD-10International Classification of Diseases 10
ORodds ratio
HRhazard ratio
HNSCChead and neck squamous cell carcinoma
RRrisk ratio
UICCUnion Internationale Contre le Cancer
WHOWorld Health Organization

References

  1. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef]
  2. Parmar, K.; Mohamed, A.; Vaish, E.; Thawani, R.; Cetnar, J.; Thein, K. Immunotherapy in head and neck squamous cell carcinoma: An updated review. Cancer Treat. Res. Commun. 2022, 33, 100649. [Google Scholar] [CrossRef] [PubMed]
  3. Burtness, B.; Harrington, K.J.; Greil, R.; Soulières, D.; Tahara, M.; de Castro, G., Jr.; Psyrri, A.; Basté, N.; Neupane, P.; Bratland, A.; et al. Pembrolizumab alone or with chemotherapy versus cetuximab with chemotherapy for recurrent or metastatic squamous cell carcinoma of the head and neck (KEYNOTE-048): A randomised, open-label, phase 3 study. Lancet 2019, 394, 1915–1928. [Google Scholar] [CrossRef] [PubMed]
  4. Cohen, E.E.W.; Soulières, D.; Le Tourneau, C.; Dinis, J.; Licitra, L.; Ahn, M.-J.; Soria, A.; Machiels, J.-P.; Mach, N.; Mehra, R.; et al. Pembrolizumab versus methotrexate, docetaxel, or cetuximab for recurrent or metastatic head-and-neck squamous cell carcinoma (KEYNOTE-040): A randomised, open-label, phase 3 study. Lancet 2019, 393, 156–167. [Google Scholar] [CrossRef]
  5. Ferris, R.L.; Blumenschein, G., Jr.; Fayette, J.; Guigay, J.; Colevas, A.D.; Licitra, L.; Harrington, K.; Kasper, S.; Vokes, E.E.; Even, C.; et al. Nivolumab for Recurrent Squamous-Cell Carcinoma of the Head and Neck. N. Engl. J. Med. 2016, 375, 1856–1867. [Google Scholar] [CrossRef] [PubMed]
  6. Borcoman, E.; Marret, G.; Le Tourneau, C. Paradigm Change in First-Line Treatment of Recurrent and/or Metastatic Head and Neck Squamous Cell Carcinoma. Cancers 2021, 13, 2573. [Google Scholar] [CrossRef]
  7. Sahu, M.; Suryawanshi, H. Immunotherapy: The future of cancer treatment. J. Oral Maxillofac. Pathol. 2021, 25, 371. [Google Scholar]
  8. Harrington, K.J.; Burtness, B.; Greil, R.; Soulières, D.; Tahara, M.; de Castro, G.; Psyrri, A.; Brana, I.; Basté, N.; Neupane, P.; et al. Pembrolizumab With or Without Chemotherapy in Recurrent or Metastatic Head and Neck Squamous Cell Car-cinoma: Updated Results of the Phase III KEYNOTE-048 Study. J. Clin. Oncol. 2023, 41, 790–802. [Google Scholar] [CrossRef]
  9. Singh, P.; McDonald, L.; Waldenberger, D.; Welslau, M.; Heyde, E.; Gauler, T.; Dietz, A. Pooled analysis of nivolumab treatment in patients with recurrent/metastatic squamous cell carcinoma of the head and neck in the United States and Germany. Head Neck 2021, 43, 3540–3551. [Google Scholar] [CrossRef]
  10. Hirsch, L.; Zitvogel, L.; Eggermont, A.; Marabelle, A. PD-Loma: A cancer entity with a shared sensitivity to the PD-1/PD-L1 pathway blockade. Br. J. Cancer 2019, 120, 3–5. [Google Scholar] [CrossRef]
  11. De Roest, R.H.; Mes, S.W.; Poell, J.B.; Brink, A.; Van De Wiel, M.A.; Bloemena, E.; Thai, E.; Poli, T.; Leemans, C.R.; Brakenhoff, R.H. Molecular Characterization of Locally Relapsed Head and Neck Cancer after Concomitant Chemoradiotherapy. Clin. Cancer Res. 2019, 25, 7256–7265. [Google Scholar] [CrossRef] [PubMed]
  12. Weber, P.; Künstner, A.; Hess, J.; Unger, K.; Marschner, S.; Idel, C.; Ribbat-Idel, J.; Baumeister, P.; Gires, O.; Walz, C.; et al. Therapy-Related Transcriptional Subtypes in Matched Primary and Recurrent Head and Neck Cancer. Clin. Cancer Res. 2022, 28, 1038–1052. [Google Scholar] [CrossRef] [PubMed]
  13. Schoenfeld, J.D.; Hanna, G.J.; Jo, V.Y.; Rawal, B.; Chen, Y.H.; Catalano, P.S.; Lako, A.; Ciantra, Z.; Weirather, J.L.; Criscitiello, S.; et al. Neoadjuvant Nivolumab or Nivolumab Plus Ipilimumab in Untreated Oral Cavity Squamous Cell Carcinoma: A Phase 2 Open-Label Randomized Clinical Trial. JAMA Oncol. 2020, 6, 1563–1570. [Google Scholar] [CrossRef] [PubMed]
  14. Uppaluri, R.; Campbell, K.M.; Egloff, A.M.; Zolkind, P.; Skidmore, Z.L.; Nussenbaum, B.; Paniello, R.C.; Rich, J.T.; Jackson, R.; Pipkorn, P.; et al. Neoadjuvant and Adjuvant Pembrolizumab in Resectable Locally Advanced, Human Papillomavirus-Unrelated Head and Neck Cancer: A Multicenter, Phase 2 Trial. Clin. Cancer Res. 2020, 26, 5140–5152. [Google Scholar] [CrossRef] [PubMed]
  15. Vos, J.L.; Elbers, J.B.W.; Krijgsman, O.; Traets, J.J.H.; Qiao, X.; van der Leun, A.M.; Lubeck, Y.; Seignette, I.M.; Smit, L.A.; Willems, S.M.; et al. Neoadjuvant immunotherapy with nivolumab and ipilimumab induces major pathological responses in patients with head and neck squamous cell carcinoma. Nat. Commun. 2021, 12, 7348. [Google Scholar] [CrossRef]
  16. Routy, B.; le Chatelier, E.; DeRosa, L.; Duong, C.P.M.; Alou, M.T.; Daillère, R.; Fluckiger, A.; Messaoudene, M.; Rauber, C.; Roberti, M.P.; et al. Gut microbiome influences efficacy of PD-1–based immunotherapy against epithelial tumors. Science 2018, 359, 91–97. [Google Scholar] [CrossRef]
  17. Zitvogel, L.; Ma, Y.; Raoult, D.; Kroemer, G.; Gajewski, T.F. The microbiome in cancer immunotherapy: Diagnostic tools and therapeutic strategies. Science 2018, 359, 1366–1370. [Google Scholar] [CrossRef]
  18. Dempsey, J.L.; Cui, J.Y. Microbiome Is a Functional Modifier of P450 Drug Metabolism. Curr. Pharmacol. Rep. 2019, 5, 481–490. [Google Scholar] [CrossRef]
  19. Weersma, R.K.; Zhernakova, A.; Fu, J. Interaction between drugs and the gut microbiome. Gut 2020, 69, 1510–1519. [Google Scholar] [CrossRef]
  20. Elkrief, A.; Derosa, L.; Kroemer, G.; Zitvogel, L.; Routy, B. The negative impact of antibiotics on outcomes in cancer patients treated with immunotherapy: A new independent prognostic factor? Ann. Oncol. 2019, 30, 1572–1579. [Google Scholar] [CrossRef]
  21. Fessler, J.; Matson, V.; Gajewski, T.F. Exploring the emerging role of the microbiome in cancer immunotherapy. J. Immunother. Cancer 2019, 7, 108. [Google Scholar] [CrossRef] [PubMed]
  22. Mura, C.; Preissner, S.; Nahles, S.; Heiland, M.; Bourne, P.E.; Preissner, R. Real-world evidence for improved outcomes with histamine antagonists and aspirin in 22,560 COVID-19 patients. Signal Transduct. Target. Ther. 2021, 6, 267. [Google Scholar] [CrossRef] [PubMed]
  23. Takiishi, T.; Fenero, C.I.M.; Câmara, N.O.S. Intestinal barrier and gut microbiota: Shaping our immune responses throughout life. Tissue Barriers 2017, 5, e1373208. [Google Scholar] [CrossRef] [PubMed]
  24. Wastyk, H.C.; Fragiadakis, G.K.; Perelman, D.; Dahan, D.; Merrill, B.D.; Yu, F.B.; Topf, M.; Gonzalez, C.G.; Van Treuren, W.; Han, S.; et al. Gut-microbiota-targeted diets modulate human immune status. Cell 2021, 184, 4137–4153.e14. [Google Scholar] [CrossRef] [PubMed]
  25. Andrews, M.C.; Duong, C.P.M.; Gopalakrishnan, V.; Iebba, V.; Chen, W.-S.; Derosa, L.; Khan, A.W.; Cogdill, A.P.; White, M.G.; Wong, M.C.; et al. Gut microbiota signatures are associated with toxicity to combined CTLA-4 and PD-1 blockade. Nat. Med. 2021, 27, 1432–1441. [Google Scholar] [CrossRef]
  26. Gopalakrishnan, V.; Helmink, B.A.; Spencer, C.N.; Reuben, A.; Wargo, J.A. The Influence of the Gut Microbiome on Cancer, Immunity, and Cancer Immunotherapy. Cancer Cell 2018, 33, 570–580. [Google Scholar] [CrossRef]
  27. Gopalakrishnan, V.; Spencer, C.N.; Nezi, L.; Reuben, A.; Andrews, M.C.; Karpinets, T.V.; Prieto, P.A.; Vicente, D.; Hoffman, K.; Wei, S.C.; et al. Gut microbiome modulates response to anti–PD-1 immunotherapy in melanoma patients. Science 2018, 359, 97–103. [Google Scholar] [CrossRef]
  28. Li, W.; Deng, Y.; Chu, Q.; Zhang, P. Gut microbiome and cancer immunotherapy. Cancer Lett. 2019, 447, 41–47. [Google Scholar] [CrossRef]
  29. Roviello, G.; Iannone, L.F.; Bersanelli, M.; Mini, E.; Catalano, M. The gut microbiome and efficacy of cancer immunotherapy. Pharmacol. Ther. 2021, 231, 107973. [Google Scholar] [CrossRef]
  30. Sepich-Poore, G.D.; Zitvogel, L.; Straussman, R.; Hasty, J.; Wargo, J.A.; Knight, R. The microbiome and human cancer. Science 2021, 371, eabc4552. [Google Scholar] [CrossRef]
  31. Iizumi, T.; Battaglia, T.; Ruiz, V.; Perez Perez, G.I. Gut Microbiome and Antibiotics. Arch. Med. Res. 2017, 48, 727–734. [Google Scholar] [CrossRef] [PubMed]
  32. Lange, K.; Buerger, M.; Stallmach, A.; Bruns, T. Effects of Antibiotics on Gut Microbiota. Dig. Dis. 2016, 34, 260–268. [Google Scholar] [CrossRef] [PubMed]
  33. Reyman, M.; van Houten, M.A.; Watson, R.L.; Chu, M.L.J.N.; Arp, K.; de Waal, W.J.; Schiering, I.; Plötz, F.B.; Willems, R.J.L.; van Schaik, W.; et al. Effects of early-life antibiotics on the developing infant gut microbiome and resistome: A randomized trial. Nat. Commun. 2022, 13, 893. [Google Scholar] [CrossRef] [PubMed]
  34. Al-Qadami, G.; Bowen, J.; Van Sebille, Y.; Secombe, K.; Dorraki, M.; Verjans, J.; Wardill, H.; Le, H. Baseline gut microbiota composition is associated with oral mucositis and tumour recurrence in patients with head and neck cancer: A pilot study. Support. Care Cancer 2023, 31, 98. [Google Scholar] [CrossRef]
  35. Nenclares, P.; Bhide, S.A.; Sandoval-Insausti, H.; Pialat, P.; Gunn, L.; Melcher, A.; Newbold, K.; Nutting, C.M.; Harrington, K.J. Impact of antibiotic use during curative treatment of locally advanced head and neck cancers with chemotherapy and radiotherapy. Eur. J. Cancer 2020, 131, 9–15. [Google Scholar] [CrossRef]
  36. Yu, Y.; Zheng, P.; Gao, L.; Li, H.; Tao, P.; Wang, D.; Ding, F.; Shi, Q.; Chen, H. Effects of Antibiotic Use on Outcomes in Cancer Patients Treated Using Immune Checkpoint Inhibitors: A Systematic Review and Meta-Analysis. J. Immunother. 2020, 44, 76–85. [Google Scholar] [CrossRef]
  37. Anthony, W.E.; Wang, B.; Sukhum, K.V.; D’souza, A.W.; Hink, T.; Cass, C.; Seiler, S.; Reske, K.A.; Coon, C.; Dubberke, E.R.; et al. Acute and persistent effects of commonly used antibiotics on the gut microbiome and resistome in healthy adults. Cell Rep. 2022, 39, 110649. [Google Scholar] [CrossRef]
  38. Shanahan, F. Changing the narrative on antibiotics. Gut 2015, 64, 1674–1675. [Google Scholar] [CrossRef]
  39. Huang, C.; Shi, G. Smoking and microbiome in oral, airway, gut and some systemic diseases. J. Transl. Med. 2019, 17, 225. [Google Scholar] [CrossRef]
  40. Langenau, J.; Boeing, H.; Bergmann, M.M.; Nöthlings, U.; Oluwagbemigun, K. The Association between Alcohol Consumption and Serum Metabolites and the Modifying Effect of Smoking. Nutrients 2019, 11, 2331. [Google Scholar] [CrossRef]
  41. Shahi, F.; Redeker, K.; Chong, J. Rethinking antimicrobial stewardship paradigms in the context of the gut microbiome. JAC-Antimicrob. Resist. 2019, 1, dlz015. [Google Scholar] [CrossRef] [PubMed]
  42. Han, C.; Song, J.; Hu, J.; Fu, H.; Feng, Y.; Mu, R.; Xing, Z.; Wang, Z.; Wang, L.; Zhang, J.; et al. Smectite promotes probiotic biofilm formation in the gut for cancer immunotherapy. Cell Rep. 2021, 34, 108706. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Modified Consort flow diagram. ICD-10: International Classification of Diseases 10, C00-14: malignant neoplasms of lip, oral cavity and pharynx.
Figure 1. Modified Consort flow diagram. ICD-10: International Classification of Diseases 10, C00-14: malignant neoplasms of lip, oral cavity and pharynx.
Cancers 15 02342 g001
Figure 2. Treatment pathways for cohort 2 (with antibiotics). The left side shows the ten most administered treatments, and the right side shows six lines of treatment (LOT). Extended spectrum penicillins (ESPs).
Figure 2. Treatment pathways for cohort 2 (with antibiotics). The left side shows the ten most administered treatments, and the right side shows six lines of treatment (LOT). Extended spectrum penicillins (ESPs).
Cancers 15 02342 g002
Table 1. Propensity score density function (purple: cohort I, green: cohort II).
Table 1. Propensity score density function (purple: cohort I, green: cohort II).
Propensity Score Density Function—Before (Left) and after (Right) Matching (Cohort I—Purple, Cohort II—Green)
Cancers 15 02342 i001Cancers 15 02342 i002
Table 2. Characteristics of cohorts I (individuals with ICD-10 codes C00-14 and no antibiotics) and II (with antibiotics) before and after matching for age and sex, BMI, secondary lymph node metastases, tobacco use and feeding device. ICD-10 = International Classification of Diseases.
Table 2. Characteristics of cohorts I (individuals with ICD-10 codes C00-14 and no antibiotics) and II (with antibiotics) before and after matching for age and sex, BMI, secondary lymph node metastases, tobacco use and feeding device. ICD-10 = International Classification of Diseases.
Before MatchingAfter Matching
Patients (n)Cohort ICohort IIp-ValueStandardized Mean DifferenceCohort ICohort IIp-ValueStandardized Mean Difference
Total21491502 13621362
Age at
index
65.764.0<0.0010.14164.564.40.7730.011
Standard deviation12.112.2 12.112.1
Males1579
73.5%
1117
74.4%
0.5460.0201043
76.6%
1007
73.9%
0.1100.061
Females570
26.5%
385
25.6%
0.5460.020319
23.4%
355
26.1%
0.1100.061
BMI24.823.9<0.0010.14824.624.10.0650.086
Standard deviation5.65.4 5.45.4
Metastases1126
31.8%
890
40.1%
<0.0010.138785
57.6%
788
57.9%
0.9070.004
Tobacco use684
32.1%
603
35.4%
0.0590.070524
38.5%
523
38.4%
0.9690.002
Feeding
device
363
16.9%
423
28.1%
<0.0010.138319
23.5%
322
23.6%
0.9610.002
Severe
sepsis
41
1.9%
145
9.7%
<0.0010.33740
2.9%
43
3.2%
0.7380.013
Table 3. Kaplan–Meier survival analysis for one year (purple: cohort I, green: cohort II).
Table 3. Kaplan–Meier survival analysis for one year (purple: cohort I, green: cohort II).
CohortPatients in CohortPatients with OutcomeMedian Survival (days)Survival Probability at End of Time Window
Iw/o antibiotics1362346--68.42%
IIw antibiotics1362511--53.89%
χ2dfp
Log-rank test61.52210.000
Hazard Ratio95% CIχ2dfp
Hazard ratio and proportionality0.58(0.51, 0.67)6.5210.011
Cancers 15 02342 i003
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Preissner, S.; Heiland, M.; Preissner, R.; Wirth, M.; Wollenberg, B. Antibiotics Significantly Decrease the Survival of Head and Neck Carcinoma Patients with Immunotherapy: A Real-World Analysis of More Than 3000 Cases. Cancers 2023, 15, 2342. https://doi.org/10.3390/cancers15082342

AMA Style

Preissner S, Heiland M, Preissner R, Wirth M, Wollenberg B. Antibiotics Significantly Decrease the Survival of Head and Neck Carcinoma Patients with Immunotherapy: A Real-World Analysis of More Than 3000 Cases. Cancers. 2023; 15(8):2342. https://doi.org/10.3390/cancers15082342

Chicago/Turabian Style

Preissner, Saskia, Max Heiland, Robert Preissner, Markus Wirth, and Barbara Wollenberg. 2023. "Antibiotics Significantly Decrease the Survival of Head and Neck Carcinoma Patients with Immunotherapy: A Real-World Analysis of More Than 3000 Cases" Cancers 15, no. 8: 2342. https://doi.org/10.3390/cancers15082342

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

Article Metrics

Back to TopTop