• Center on Health Equity and Access
  • Clinical
  • Health Care Cost
  • Health Care Delivery
  • Insurance
  • Policy
  • Technology
  • Value-Based Care

Current and Emerging Microbiome-Targeted Therapies to Prevent Recurrent CDI: A Stakeholder Interchange Report

Publication
Article
Supplements and Featured PublicationsCurrent and Emerging Microbiome-Targeted Therapies to Prevent Recurrent CDI: A Stakeholder Interchange Report

INTRODUCTION

Clostridioides difficile is a common infection associated with substantial health-related burdens. Panelists among several health care disciplines participated in a recent AJMC® Health System Stakeholder Interchange entitled, “Current and Emerging Microbiome-Targeted Therapies to Prevent Recurrent Clostridioides difficile Infection.” Moderated by Ryan Haumschild, PharmD, MS, MBA, of Emory Healthcare and Winship Cancer Institute in Atlanta, Georgia, this conversation highlighted important considerations for improving the management of C difficile infections (CDIs) and reducing the risk of relapse. The speakers focused upon understanding recurrent CDIs; differentiating among probiotics, fecal microbiota transplant (FMT), and live biotherapeutic products (LBPs); and grasping the economic and clinical impacts of this difficult medical condition.

BACKGROUND

The microbiota are a community of microbiomes. The microbiome includes microorganisms such as fungi, bacteria, and viruses that inhabit a specific environment. Firmicutes and Bacteroides phyla represent approximately 90% of colonial gut microorganisms.1 These phyla play protective roles in pathogen resistance, immune response, and injury protection, and they are involved in the biosynthesis and metabolism of drugs, nutrients, and energy.

Factors contributing to CDI in the gut include advanced age, medication usage, and a suboptimal diet, which can significantly disrupt natural eubiosis and lead to a notable reduction in microbial diversity. A higher level of microbial diversity within the gut is associated with a healthier gut environment. In contrast, decreased diversity results in the loss of beneficial microbes and an increase in pathogenic microbes such as C difficile. This microbial imbalance contributes to inflammation and the increased proliferation of C difficile, which underlies the symptomatic manifestations of this gastrointestinal (GI) disease.

Dysbiosis, an imbalance of the gut microbiota, plays an important role in the development of CDI and particularly in recurrent infections. The pathogenic process of CDIs involves a crucial step of colonizing gut cells, contingent upon the contest between specific colonization factors produced by C difficile and colonization resistance (barrier effect) exerted by the gut microbiota.2 Certain factors (eg, use of antimicrobial therapy) may lead to the elimination of the protective effect of commensal bacteria; the subsequent availability of previously inaccessible niches then makes the intestine susceptible to C difficile colonization.3 These recurrent infections pose a serious health care–associated risk and account for approximately 75,000 to 175,000 additional cases of CDI per year in the United States.4 The inability to effectively treat these infections has serious implications that include decreased quality of life (QOL), increased hospitalizations, and fatalities.

CLINICAL IMPACT OF RECURRENT C DIFFICILE INFECTION

Recurrence in Hospitalized Patients

Dr Feuerstadt reviewed the crucial nature of recognizing the broader burden of disease associated with CDIs and complications that extend beyond abdominal pain and diarrhea to cause severe consequences (eg, sepsis, need for colectomy). Results of outcome studies involving commercially insured individuals demonstrated an increased occurrence of sepsis and colectomy with each recurrent infection. Recurrence rates of 15% to 30% overall have been reported in patients who initially responded to antimicrobial therapy, and they have reached approximately 45% to 65% after the second infection.5 Dr Feuerstadt added that the attributable mortality rate associated with CDI is approximately 16.4% for the initial occurrence and 39% for subsequent occurrences. A significant impact on health-related QOL (HRQOL) may even result in post-traumatic stress disorder among affected patients who are given suitable treatment but who remain at risk of recurrence.

The patients whom Dr Chopra encounters in the hospital setting, particularly those arriving from long-term care facilities and nursing homes, exhibit a complex clinical profile. She stated, “They have a history of exposure to antibiotics, multiple medical devices, and multiple comorbidities.” Upon presentation, they may manifest severe hypertension, sepsis, and other signs of critical illness with recurrent CDI. It is not uncommon for these patients to have concomitant infections, including those caused by multidrug-resistant organisms. Their overall health status is greatly compromised due to chronic dysbiosis. These patients pose a significant concern due to their elevated morbidity and mortality rates. Addressing the dysbiosis they experience is imperative to improve outcomes for this vulnerable population.

Importantly, Dr Feuerstadt noted, the risk of having another CDI increases after a patient experiences a recurrence. He also emphasized the importance of considering the broader context of individuals who are more susceptible to recurrence and the evolving epidemiology of the bacteria itself. The NAP1/B1/O27 C difficile strain, known for its hypervirulence, played an important role in the initial CDI pandemic around the world.5 Currently, the possible lower prevalence of this strain may indicate a decline in its virulence. He mentioned an alternate viewpoint—still other strains that may be less detectable and identifiable using current testing methods may contribute to recurrence, particularly in more susceptible populations with inflammatory bowel disease (IBD).

Despite these multiple factors associated with recurrence, health care systems have made significant progress. According to Dr Feuerstadt, data from the Emerging Infections Program showed a decrease in health care–associated infection from approximately 66% in 2011 to about 50% in 2017. This improvement can be attributed to implementation of better infection control measures by health care systems, stricter antimicrobial usage policies led by infectious disease professionals, and overall enhanced treatment strategies.

In addition to patient characteristics, the aggregate recurrence rate after 3 or more episodes is approximately 40% across different patient profiles, according to Dr Khanna. He also stated that the results of trials evaluating SER-109, CP101, and RBX 2660 showed a success rate of 58%. Thus, use of newer guidelines may provide more refined treatment strategies, such as upfront use of fidaxomicin, treatment with bezlotoxumab, and elimination of metronidazole therapy in favor of vancomycin tapers. These observations, however, are based on the results of a limited number of trials. Our evolving strategies may lead to lower recurrence rates for susceptible patients with multiple recurrences and eventually may lead to management of the underlying seed of infection.

IMPORTANCE OF EARLY DETECTION

Dr Chopra explained that CDI recurrence has had a notable impact on the QOL of patients and has caused considerable burdens for hospitals; these include increased economic costs and prolonged hospital stays. The challenge of recurrent CDIs highlights a significant gap in our knowledge regarding the epidemiology of this infection. Dr Khanna agreed, “It’s important for us to have early recognition because we have seen a shift in the epidemiology of C difficile, and we’re now seeing that bug out in the community, leading to community-acquired infection. That’s over half of all C difficile.” He also was concerned that some patients undergo multiple diagnostic studies, including colonoscopy, before a stool test for C difficile is performed, and he emphasized that clinicians must maintain a high level of suspicion for this infection beyond the hospital setting.

Timely and accurate diagnosis is most important when dealing with recurrent CDIs. Polymerase chain reaction (PCR) testing for C difficile can identify the toxin genes directly in stool specimens and other samples.6 This highly sensitive and specific method for detecting the organism is considered to be a popular rapid test for C difficile diagnosis.6 PCR allows for early detection of C difficile and provides rapid results.6 Staff at Dr Chopra’s health care center have transitioned from using PCR as the primary diagnostic tool to employing a 2-step antigen and antibody test proven to be highly effective. This 2-step test for CDI is a diagnostic approach that detects specific antigens and antibodies associated with CDI.7 In the first step, immunoassays are used to test patient samples (typically, stool specimens) for the presence of C difficile toxins or antigens.7 A positive result indicates the presence of toxins and an active CDI.7 If the antigen test is positive or there is a clinical suspicion of CDI, the second step involves testing the patient’s blood sample for the presence of C difficile–specific antibodies.7 Antibody tests help determine previous exposure or current infection. Rising antibody levels may indicate an active infection, whereas the presence of specific antibodies suggests past exposure to the bacterium.7 When she receives inconclusive results for both tests, Dr Chopra proceeds with reflex testing to PCR. This comprehensive approach ensures that health care providers make accurate diagnoses. Thus, diagnosis of recurrent CDI requires clinical acumen, and it must extend testing alone.

Dr Khanna fully endorsed the testing modality described by Dr Chopra, particularly the use of a multistep assay, which is considered to be the ideal test. Dr Khanna stated, “However, it is important to acknowledge that the best test is the one that is readily available and feasible to perform.” He also emphasized that according to recent data, approximately 75% to 80% of laboratories perform the PCR assay as a stand-alone test for C difficile. Use of this approach requires that health care professionals exercise clinical acumen and ensure that the patient’s clinical presentation aligns with the diagnostic criteria. Clinical acumen must be combined with a comprehensive patient history, assessment of risk factors and symptoms associated with C difficile, and appropriate diagnostic tests. When there is still uncertainty or multiple potential causes of diarrhea, a positive PCR result may indicate CDI, although other contributing factors cannot be ruled out.

The importance of early diagnosis cannot be overstated, particularly from the standpoint of a provider who manages patients with IBD. Delayed diagnosis of concomitant CDI can exacerbate IBD. Both patients and providers may find differentiation of a flare related to IBD or CDI to be difficult without proper testing. Dr Khanna stated, “Providers need to demonstrate clinical assessment skills and curiosity. Instead of assuming that a flare is solely related to IBD, they must consider the possibility of CDI or other infections.”

Treatment options for CDI can also help health care providers to recognize active IBD. Use of oral vancomycin or fidaxomicin primarily targets CDI alone; fidaxomicin does not address any other conditions, and physicians are prompted to reevaluate the situation and consider alternative diagnoses if a patient fails to show improvement with these treatments.

Dr Abraham explained that reassessment of patients becomes crucial within the population, since traditional risk factors for CDI do not necessarily apply to patients with IBD. These patients are typically younger, and they may not have a prior hospitalization history. He noted that, surprisingly, there was more than a 2-fold increased risk of hospitalization was noted after a patient was diagnosed with CDIs.8 To curtail costs, early diagnosis must be achieved, and treatment must be initiated promptly and before patients require hospitalization. In many cases, these patients are not exposed to antibiotics before infection. Instead, dysbiosis that is often intertwined with ongoing active inflammation of IBD appears to play a role. Early diagnosis enables effective management of CDI and eases assessment to ensure that the symptoms are not solely attributed to IBD disease activity. Appropriate treatment strategies are essential to avoid unnecessary costs, prevent the potential exacerbation of both conditions, and maintain a balanced approach to patient care.

TREATMENT OPTIONS

For first recurrence of CDI, preferred treatment options include use of fidaxomicin or an alternative therapy (eg, vancomycin) administered orally in a tapered or pulsed manner. In the event of a second or subsequent recurrence, treatment options may involve fidaxomicin, vancomycin, or fecal transplant. Administration of at least 2 antibiotic treatments for each recurrent infection is recommended before FMT is considered. Additionally, use of monoclonal antibodies (eg, bezlotoxumab) can be considered; this may be administered as an intravenous infusion given once over 1 hour and only for recurrent CDI.9

ROLE OF ANTIBIOTICS IN THE TREATMENT OF RECURRENT C DIFFICILE

Antibiotic therapy is essential when treating initial acute episodes of CDI. For management of recurrent CDI, the question arises of whether to use antibiotics alone or to combine them with a recurrence prevention strategy, noted Dr Khanna. Dr Feuerstadt highlighted that the infection has 2 components: the vegetative phase is primarily targeted with antibiotics, and the spore phase can be effectively managed by restoring the microbiota. When disrupted microbial diversity is not restored, the chance of recurrence increases. In some cases, recurrence prevention strategies can be incorporated within the antibiotic regimen itself. For example, a taper or pulse regimen can be employed with vancomycin; alternatively, a fidaxomicin pulse regimen as demonstrated in the EXTEND protocol may be used.10 By incorporating supplementation or restoration of the microbiota, we can significantly reduce recurrence rates. This comprehensive approach that combines antimicrobial therapy with microbiota restoration helps to address both aspects of the infection and to improve the overall effectiveness of the treatment.

Dr Chopra added that the initial antibiotic used in the first episode can impact subsequent treatment options. For example, if metronidazole is given initially, switching to vancomycin or fidaxomicin may be preferred to treat the recurrence. Fidaxomicin therapy has been associated with reduced recurrence rates.10 However, individual patient factors, such as benefit coverage and cost, that may influence the choice of antibiotics are important. Further, not all patients have access to fidaxomicin. Clinical, practical, and cost-related factors all must be considered.

Bezlotoxumab, an FDA-approved monoclonal antibody, actively targets the toxin produced by C difficile; its use is intended specifically for recurrence prevention in CDI.9 The results of MODIFY I and MODIFY II (NCT01241552 and NCT01513239) demonstrated its ability to decrease the overall risk of recurrence by approximately 10%, although its effectiveness may vary depending upon specific risk factors.11 The utility of bezlotoxumab is limited and not universal, but the drug has been available as a viable option for approximately 6 years, and its use is warranted in select cases. The indication for bezlotoxumab focuses on reducing CDI recurrence in patients 18 years and older who currently receive antibacterial drug treatment for CDI and who are identified as high-risk for recurrence.9 Importantly, bezlotoxumab is not intended for the treatment of CDI itself, and it does not possess antibacterial properties.9

Comparing and Contrasting Live Biotherapeutic Products, FMT, and Probiotics

LBPs are defined by the FDA as biological products containing live organisms; they are distinct from vaccines and are specifically designed to prevent, treat, or potentially cure diseases.12 One example is fecal microbiota, live-jslm (Rebyota; Ferring Pharmaceuticals), which was approved by the FDA in November 2022.13 It consists of live fecal microbiota that are used to treat recurrent CDI in adults following antibiotic therapy. Each 150-mL dose of this suspension contains approximately 1 × 108 to 5 × 1010 colony-forming units (CFU)/mL of fecal microbes, including more than 1 × 105 CFU/mL of Bacteroides.14 The patient must be carefully screened before receiving a 1-time, 150-mL dose of the suspension delivered as an enema. The administration process involves several important considerations, including having a suitable setting for its use and identifying appropriate health care professionals responsible for its administration. Clinical trial data supporting approval this product have generated significant interest.

Another LBP known as fecal microbiota spores, live-brpk (Vowst; Seres Therapeutics/Nestle Health Science) was approved by the FDA in April 2023 to prevent recurrent CDI in adults who have experienced at least 1 prior antibiotic treatment.15 This product is available as oral capsules that contain 1 × 106 to 3 × 107 Firmicutes spore CFU from healthy, screened stool from human donors. It is given as 4 capsules taken orally for 3 consecutive days.16

These 2 therapeutic options offer new possibilities that were previously unavailable. Importantly, the diverse treatment options also feature different delivery mechanisms for patients. Long-term efficacy is a critical aspect of interest for both payer colleagues and clinicians. Recurrent CDIs impose a substantial burden on patients, further emphasizing the need for effective long-term solutions.

Dr Khanna noted that because LBPs do not adhere to the conventional definition of probiotics, their categorization as probiotics is inappropriate. Production of probiotics is not strictly governed by a requirement for live bacteria, and the designation is not contingent upon alignment with specific regulatory guidelines. One intervention involves the ingestion of bacterial spores with their associated nutrient source rather than of live bacteria. The FDA considers the viability of microorganisms to be important for regulatory approval, but practical clinicians typically prioritize treatment decisions based upon empirical evidence rather than rigid definitions.

Live-brpk and live-jslm play distinct roles in managing patients diagnosed with recurrent CDIs. Some patients may decline live-jslm due to their aversion to rectal administration, whereas others may prefer the convenience of oral tablets provided by fecal microbiota, live-brpk. Whereas supporting clinical data are available for the oral tablets, more comprehensive and prolonged data are available for the rectal product. Further, there are notable disparities between the products. Fecal microbiota, live-jslm encompasses a broader range of live bacteria and presents a wider spectrum of bacterial diversity, whereas fecal microbiota, live-brpk has a narrower composition. Logistically, fecal microbiota, live-jslm administration necessitates the services of a health care provider and a clinic equipped to administer the enema. Conversely, fecal microbiota, live-brpk can be prescribed and taken in various settings, allowing greater flexibility. Therefore, when determining the appropriate treatment option for individual patients, logistical challenges must be taken into account. Importantly, fecal microbiota, live-jslm and live-brpk are distinct products, and long-term data must be analyzed to fully comprehend their efficacy and safety profiles. Continued monitoring and observation will provide insights for the optimal use of these treatments for patients with recurrent CDI.

FECAL TRANSPLANT

Historically, fecal transplantation has played a significant role in addressing CDIs. This procedure involves instilling donor feces through an enema to promote the transition from dysbiosis to eubiosis in patients with recurrent infections. The results of observational studies have shown an efficacy rate of nearly 90% for fecal transplant.17 However, this procedure entails 2 essential considerations. First, the procedure must be performed exclusively within a health care facility by a qualified health care professional. Second, the fecal matter must be sourced from a reputable stool bank or a health care facility to ensure adherence to stringent standards and protocols. Donor screening is a critical step that involves numerous tests and ensures the suitability of a stool sample. The treatment process typically includes antibiotic therapy followed by a washout period of 1 to 2 days to comply with the treatment protocol. Bowel preparation is then performed before the fecal transplant; in numerous cases, administration via the lower GI route has been more effective.

ADVANTAGES OF LBP VS FMT

Compared with conventional fecal transplant procedures performed via colonoscopy, LBPs offer noninvasive therapies that eliminate the need for anesthesia and sedation. However, the most significant advantage lies in the standardization and traceability of the supply chain, which ensures that the source can be identified and addressed in the rare case that an adverse event (AE) occurs. In contrast, AEs related to conventional fecal transplant may go unreported or become challenging to trace back to the specific donor. When 1 donor provides material to multiple recipients without traceability, AEs may propagate and affect multiple individuals without detection. These advantages underscore the importance of standardized and traceable supply chains in the context of FMTs.

ROLE OF PROBIOTICS

Probiotics are frequently used in the management of CDIs, but guidelines typically do not endorse their use.18 Nonetheless, understanding of potential mechanisms of action can shed light on the role of probiotics in managing recurrent CDIs. First, probiotics may promote a competitive environment within the gut by competing for available resources. Second, they can enhance the crucial function of the gut barrier. Third and last, probiotics may exert their effects through immune modulation. Among probiotics commonly studied, Lactobacillus and other strains have demonstrated stronger efficacy. However, probiotics likely are not effective as a primary treatment for CDI.

ECONOMIC IMPACT

As the panelists delved into the economic impact of CDI, their considerations transcended frontline therapy and encompassed various factors, including potential hospitalizations and increased use of health care resources. In 2016, the annual cost associated with CDI in the United States was approximately $6.3 billion.19 The costs related to recurrent infections are mainly driven by hospitalizations—each patient with a CDI may require an additional hospital stay that may last just 3 days to as long as 20 days.4

The Diagnosis-Related Group (DRG) system, a health care classification system, groups patients based upon their clinical conditions and treatment needs.20 Dr Evans reported that Medicare patients with the relevant DRG had average hospitalization costs of approximately $38,000, with the costs for Medicaid patients potentially being lower. However, Dr Evans also acknowledged that patients experiencing severe complications such as sepsis, admission to an intensive care unit, and intubation could face costs reaching hundreds of thousands of dollars. This system ensures fair and consistent reimbursement for hospitals and other health care providers while promoting efficiency in health care delivery.

The estimated cost to manage an initial episode of CDI in commercially insured inpatient populations is approximately $71,980, according to Dr Feuerstadt. However, he noted that when the infection recurs and reaches the second occurrence and beyond, the cost dramatically increases to around $207,733.4

PAYER’S PERSPECTIVE

Dr Feuerstadt emphasized the significance of considering payers to be key stakeholders and incorporating their insights on these new treatment options. To ensure a comprehensive understanding and collaborative decision-making process, payers must be stakeholders in the ongoing conversation. As Dr Khanna emphasized, the paramount consideration is ensuring that patients can readily obtain the most suitable product for their needs. This underscores the significance of accessibility and availability when the efficacy of a treatment option is evaluated.

The recent approval of fecal microbiota, live-brpk introduces a new dynamic in the market, and its evaluation by payers may take time. Typically, during the initial 6 months after approval, access to a new treatment may be restricted as its efficacy undergoes a review by the pharmacy and therapeutics committee of a hospital. However, exceptions can be made through a peer-to-peer review that presents the case for patient-specific needs and allows for more efficient resolution.

For Medicare patients, the oral agent fecal microbiota, live-brpk falls under Part D of the benefit design, whereas fecal microbiota, live-jslm is categorized under Part B. This distinction has implications, especially considering the implementation of the Inflation Reduction Act policy earlier this year. The benefit design is important, because it determines the cost-sharing arrangements for patients. Medicare patients, for instance, may face significant financial challenges due to co-insurance requirements; Dr Lopes cautioned that co-insurance costs could be as high as 20% to 30%, depending upon the specific plan. Unlike coverage by commercial insurance, Medicare coverage does not allow use of prescription discount cards, coupons, or patient assistance programs that are made available by manufacturers. However, foundation programs may provide some financial support to referred patients. On the other hand, under Medicare Part B, costs are typically limited; any co-insurance involved is usually set at 20%. The overall cost structure can significantly impact patient affordability, and it may serve as a limiting factor for certain individuals.

Another important consideration for payers is the predictability of cost and the consistency associated with the use of fecal transplant therapies. The concept of establishing specialized centers for fecal transplant—like those for heart, kidney, or liver grafts—can provide enhanced predictability and consistency in terms of outcomes and costs. This approach allows for a higher level of actuarial comfort for payers, as it ensures standardized procedures, expertise, and quality control measures in the delivery of fecal transplant therapies. With procedures centralized within specialized facilities, payers can expect a more reliable and consistent approach that contributes to a better overall cost predictability and improved patient outcomes.

Payers have significantly changed their approach to FMT over the years. Initially, there was a higher frequency of prior authorization and preapproval requests for FMT procedures. However, as experience and evidence have accumulated, there is less need for such authorizations. Currently, prior authorization or preapproval is needed only about once every 6 months, even for high-volume centers performing a substantial annual number of FMT procedures.

The evolution of payers’ perspectives on FMT is encouraging; hopefully, this trend will continue with LBPs. The ultimate goal is to ensure that patients can access the most effective therapy available. This entails minimizing the financial burden on patients by reducing out-of-pocket costs and streamlining the approval process by having payers impose few hurdles.

The financial cost of FMT encompasses not only fecal transplant materials but colonoscopy and anesthesia. As the discussion progresses and payers consider the use of LBPs, the elimination of required colonoscopy can contribute to cost savings. This shift in perspective should be considered when evaluating the overall cost-effectiveness and feasibility of LBPs in comparison to traditional FMT procedures.

FMT-RELATED COSTS

The financial cost of traditional FMTs encompasses components that can vary across centers, states, and cities. They include expenses associated with screening, processing, and storing the fecal material and with the accompanying colonoscopy procedure. The colonoscopy cost is comprised of fees related to performing the procedure, which may include expenditures for services by the physician, an anesthesiologist, and the facility. Other costs involve patients needing to be away from work for at least 1.5 to 2 days for recovery.

Moreover, potential complications arising during the hospital stay can further impact overall expenses. Dr Khanna stated that the oral FMT formulation was assigned a price of $17,500; however, whether that referred to the average wholesale price or the consumer price was unclear. He added that the rectal formulation of FMT was currently priced at $9,000. It is reasonable to assume that there may be additional costs related to rectal administration, because health care professionals perform the procedure. Both formulations may need to be stored in a freezer until they are dispensed, which leads to storage costs.

In summary, there is a cost disparity between the 2 FMT formulations, with the oral option being more expensive. Furthermore, the cost of colonoscopy remains variable and contributes to the overall expense associated with traditional FMT procedures.

RECURRENT CDI AND QUALITY OF LIFE

CDI poses significant challenges to patients and health care systems, as it impacts HRQOL and results in substantial use of health care resources. Understanding the various dimensions of this issue, including the psychological impact on patients and the efficacy of different treatments, is essential for addressing the concerns and needs affected patients.

In a study conducted in 2020, approximately 350 patients were surveyed to assess the HRQOL associated with CDI.21 One-third of the participants had active C difficile, and the remaining two-thirds had a past medical history of the infection.21 The survey focused on the physical, emotional, adaptational, relational, and productivity impacts on patients.21 The results consistently demonstrated a negative impact across these domains for both patients with active infections and those with a distant history of the infection.21

The psychological effects on patients with CDI have been observed to cause subsequent patterns of health care use. Many patients seek frequent office visits and emergency department care to help them deal with their concerns and fears. Often, they desire microbiota restoration therapy or treatment with rituximab to reduce C difficile recurrence. Additionally, patients resort to online research to find information on potential treatments, and this often makes them question why their health care providers have not offered certain treatments. This highlights a potential gap between patient expectations and available treatment options.

The C difficile Health-Related QOL 32-Questionnaire is a pivotal tool in assessing HRQOL. This questionnaire assesses the physical, emotional, and social impacts of CDIs on patients.22 The data obtained from this questionnaire indicate that recurrent infections are associated with a decrease in HRQOL. However, treatments such as fecal microbiota, live-jslm and fecal microbiota, live-brpk provide improvement in HRQOL, whereas administration of placebo causes no change in this score.

Health care providers, researchers, and policymakers must collaborate to enhance patient outcomes, reduce the burden on health care systems, and explore innovative treatment options such as fecal microbiota, live-jslm and fecal microbiota, live-brpk. By prioritizing the well-being of individuals affected by CDI, we can work toward improving HRQOL and ultimately achieve better overall health care outcomes.

CLOSING REMARKS

Early diagnosis and timely intervention are crucial for managing CDIs, in terms of both initial diagnosis and recurrence. Treatment decisions should be based on reviewing the patient’s prior treatment history and ensuring that adherence issues are addressed before considering alternative therapies.

One exciting development in the field is the availability of LBPs, which offer a means to restore the microbiome in patients with multiple recurrences. The presence of FDA-approved and reliable LBPs provides health care providers with a promising option for improving patient outcomes. The focus now is on evaluating the long-term impact of LBPs on patients’ QOL and gathering more data to support their efficacy.

From a health care provider’s perspective, the selection of the most cost-effective medication that effectively addresses the specific situation often is preferred. In comparing fidaxomicin with vancomycin, fidaxomicin shows potential for preventing recurrences; however, the cost difference between the 2 drugs needs to be carefully considered. The financial aspect is important, but health care providers recognize the need to prioritize patient outcomes over cost. Achievement of the best possible results for patients and a focus on the relationship between quality outcomes and cost should be emphasized.

Creating best practices and streamlining care is an opportunity presented by the use of LBPs and other innovative therapies. Payers play a crucial role in assessing the appropriateness of these treatments and evaluating their impact on reducing recurrences, improving function, enhancing QOL, and managing the overall cost of care. Having choices and relying on data are important to ensure that payers are comfortable when using these treatments and to accept of their associated outcomes.

In conclusion, the management of CDIs involves early diagnosis, tailored treatment approaches, and a focus on achieving optimal results. The availability of LBPs offers a promising avenue for restoring the microbiome and improving patients’ QOL. Cost considerations are important, but delivery of the best possible patient outcome is the priority. Streamlining care practices and involving payers in the conversation contribute to the development of effective strategies and the use of treatments that demonstrate both clinical efficacy and cost effectiveness.

REFERENCE LIST

1. Rinninella E, Raoul P, Cintoni M, et al. What is the healthy gut microbiota composition? A changing ecosystem across age, environment, diet, and diseases. Microorganisms. 2019;7(1):14. doi:10.3390/microorganisms7010014

2. Péchiné S, Janoir C, Boureau H, et al. Diminished intestinal colonization by Clostridium difficile and immune response in mice after mucosal immunization with surface proteins of Clostridium difficile. Vaccine. 2007;25(20):3946-3954. doi:10.1016/j.vaccine.2007.02.055

3. De La Cochetière MF, Durand T, Lalande V, Petit JC, Potel G, Beaugerie L. Effect of antibiotic therapy on human fecal microbiota and the relation to the development of Clostridium difficile. Microb Ecol. 2008;56(3):395-402. doi:10.1007/s00248-007-9356-5

4. Burton HE, Mitchell SA, Watt M. A systematic literature review of economic evaluations of antibiotic treatments for Clostridium difficile infection. Pharmacoeconomics. 2017;35(11):1123-1140. doi:10.1007/s40273-017-0540-2

5. Song JH, Kim YS. Recurrent Clostridium difficile infection: risk factors, treatment, and prevention. Gut Liver. 2019;13(1):16-24. doi:10.5009/gnl18071

6. Bagdasarian N, Rao K, Malani PN. Diagnosis and treatment of Clostridium difficile in adults: a systematic review. JAMA. 2015;313(4):398-408. doi:10.1001/jama.2014.17103

7. Solomon DA, Milner DA Jr. ID learning unit: understanding and interpreting testing for Clostridium difficile. Open Forum Infect Dis. 2014;1(1):ofu007. doi:10.1093/ofid/ofu007

8. Khanna S. Management of Clostridioides difficile infection in patients with inflammatory bowel disease. Intest Res. 2021;19(3):265-274. doi:10.5217/ir.2020.00045

9. Zinplava. Prescribing information. Merck; 2023. Accessed June 19, 2023. https://www.merck.com/product/usa/pi_circulars/z/zinplava/zinplava_pi.pdf

10. Guery B, Menichetti F, Anttila VJ, et al. Extended-pulsed fidaxomicin versus vancomycin for Clostridium difficile infection in patients 60 years and older (EXTEND): a randomised, controlled, open- label, phase 3b/4 trial. Lancet Infect Dis. 2018;18(3):296-307. doi:10.1016/S1473-3099(17)30751-X

11. Wilcox MH, Gerding DN, Poxton IR, et al.; MODIFY I and MODIFY II Investigators. Bezlotoxumab for prevention of recurrent Clostridium difficile infection. N Engl J Med. 2017;376(4):305-317. doi:10.1056/NEJMoa1602615

12. Early clinical trials with live biotherapeutic products: chemistry, manufacturing, and control information. Guidance for industry. US Department of Health and Human Services; FDA Center for Biologics Evaluation and Research. Updated 2016. Accessed June 21, 2023. https://www.fda.gov/files/vaccines,%20blood%20%26%20biologics/published/Early-Clinical-Trials-With-Live-Biotherapeutic- Products--Chemistry--Manufacturing--and-Control-Information--Guidance-for-Industry.pdf

13. Ferring Pharmaceuticals announces availability of REBYOTA™ (fecal microbiota, live – jslm) – the first and only FDA-approved microbiome-based treatment for the prevention of recurrence of C. diff infection in adults. News release. Ferring Pharmaceuticals. February 14, 2023. Accessed June 21, 2023. https://ferringusa.com/?press=ferring-pharmaceuticals-announces-availability-of-rebyota-fecal-micro- biota-live-jslm-the-first-and-only-fda-approved-microbiome-based-treatment-for-the-prevention-of-r

14. Rebyota. Prescribing information. Ferring Pharmaceuticals. Accessed June 20, 2023.
https://www.fda.gov/media/163587/download

15. Seres Therapeutics and Nestlé Health Science announce U.S. commercial availability of VOWST™, the first and only FDA-approved microbiota-based oral therapeutic for prevention of recurrence of C. difficile infection. News release. Business Wire. June 05, 2023. Accessed June 20, 2023. https://www.businesswire.com/news/home/20230605005120/en/Seres-Therapeutics-and- Nestl%C3%A9-Health-Science-Announce-U.S.-Commercial-Availability-of-VOWST%E2%84%A2- the-First-and-Only-FDA-Approved-Microbiota-Based-Oral-Therapeutic-for-Prevention-of- Recurrence-of-C.-Difficile-Infection

16. Vowst. Prescribing information. Seres Therapeutics/Nestlé Health Science; 2023. Accessed June 20, 2023. https://www.serestherapeutics.com/our-products/VOWST_PI.pdf

17. Liubakka A, Vaughn BP. Clostridium difficile infection and fecal microbiota transplant. AACN Adv Crit Care. 2016;27(3):324-337. doi:10.4037/aacnacc2016703

18. Johnson S, Lavergne V, Skinner AM, et al. Clinical practice guideline by the Infectious Diseases Society of America (IDSA) and Society for Healthcare Epidemiology of America (SHEA): 2021 focused update guidelines on management of Clostridioides difficile infection in adults. Clin Infect Dis. 2021;73(5):e1029–e1044, doi:10.1093/cid/ciab549

19. Gupta A, Ananthakrishnan AN. Economic burden and cost-effectiveness of therapies for Clostridioides difficile infection: a narrative review. Therap Adv Gastroenterol. 2021;14:17562848211018654. doi:10.1177/17562848211018654

20. Design and development of the Diagnosis Related Group (DRG). Centers for Medicare & Medicaid Services. October 2019. Accessed June 21, 2023. https://cms.gov/icd10m/version37-fullcode-cms/fullcode_cms/Design_and_development_of_the_Diagnosis_Related_Group_(DRGs).pdf

21. Lurienne L, Bandinelli PA, Galvain T, Coursel CA, Oneto C, Feuerstadt P. Perception of quality of life in people experiencing or having experienced a Clostridioides difficile infection: a US population survey. J Patient Rep Outcomes. 2020;4(1):14. doi:10.1186/s41687-020-0179-1

22. Garey KW, Aitken SL, Gschwind L, et al. Development and validation of a Clostridium difficile health-related quality-of-life questionnaire. J Clin Gastroenterol. 2016;50(8):631-637. doi:10.1097/MCG.0000000000000473

© 2024 MJH Life Sciences
AJMC®
All rights reserved.