References

Reinke JM, Sorg H. Wound repair and regeneration. Eur Surg Res. 2012; 49:(1)35-43 https://doi.org/10.1159/000339613

Hobley L, Harkins C, MacPhee CE, Stanley-Wall NR. Giving structure to the biofilm matrix: an overview of individual strategies and emerging common themes. FEMS Microbiol Rev. 2015; 39:(5)649-669 https://doi.org/10.1093/femsre/fuv015

Bjarnsholt T. The role of bacterial biofilms in chronic infections. APMIS. 2013; 121:(136)1-58 https://doi.org/10.1111/apm.12099

Kalan L, Zhou M, Labbie M, Willing B. Measuring the microbiome of chronic wounds with use of a topical antimicrobial dressing–a feasibility study. PLoS One. 2017; 12:(11) https://doi.org/10.1371/journal.pone.0187728

Omar A, Wright J, Schultz G Microbial biofilms and chronic wounds. Microorganisms. 2017; 5:(1) https://doi.org/10.3390/microorganisms5010009

Percival SL, McCarty SM, Lipsky B. Biofilms and wounds: an overview of the evidence. Adv Wound Care. 2015; 4:(7)373-381 https://doi.org/10.1089/wound.2014.0557

Ren D, Madsen JS, Sørensen SJ, Burmølle M. High prevalence of biofilm synergy among bacterial soil isolates in cocultures indicates bacterial interspecific cooperation. ISME J. 2015; 9:(1)81-89 https://doi.org/10.1038/ismej.2014.96

Seth AK, Geringer MR, Galiano RD Quantitative comparison and analysis of species-specific wound biofilm virulence using an in vivo, rabbit-ear model. J Am Coll Surg. 2012; 215:(3)388-399 https://doi.org/10.1016/j.jamcollsurg.2012.05.028

Seth AK, Geringer MR, Hong SJ Comparative analysis of single-species and polybacterial wound biofilms using a quantitative, in vivo, rabbit ear model. PLoS One. 2012; 7:(8) https://doi.org/10.1371/journal.pone.0042897

Schultz G, Bjarnsholt T, James GA Consensus guidelines for the identification and treatment of biofilms in chronic nonhealing wounds. Wound Repair Regen. 2017; 25:(5)744-757 https://doi.org/10.1111/wrr.12590

Snyder RJ, Bohn G, Hanft J Wound biofilm: current perspectives and strategies on biofilm disruption and treatments. Wounds. 2017; 29:(6)S1-S17

Wolcott RD, Rumbaugh KP, James G Biofilm maturity studies indicate sharp debridement opens a time-dependent therapeutic window. J Wound Care. 2010; 19:(8)320-328 https://doi.org/10.12968/jowc.2010.19.8.77709

Schwartz JA, Goss SG, Facchin F Surgical debridement alone does not adequately reduce planktonic bioburden in chronic lower extremity wounds. J Wound Care. 2014; 23:(9)S4-S13 https://doi.org/10.12968/jowc.2014.23.Sup9.S4

Attinger C, Wolcott R. Clinically addressing biofilm in chronic wounds. Adv Wound Care. 2012; 1:(3)127-132 https://doi.org/10.1089/wound.2011.0333

Wolcott R. Disrupting the biofilm matrix improves wound healing outcomes. J Wound Care. 2015; 24:(8)366-371 https://doi.org/10.12968/jowc.2015.24.8.366

Kim D, Namen Ii W, Moore J Clinical assessment of a biofilm-disrupting agent for the management of chronic wounds compared with standard of care: A therapeutic approach. Wounds. 2018; 30:(5)120-130

Driver VR, Eckert KA, Carter MJ, French MA. Cost-effectiveness of negative pressure wound therapy in patients with many comorbidities and severe wounds of various etiology. Wound Repair Regen. 2016; 24:(6)1041-1058 https://doi.org/10.1111/wrr.12483

Naimark DM, Bott M, Krahn M. The half-cycle correction explained: two alternative pedagogical approaches. Med Decis Making. 2008; 28:(5)706-712 https://doi.org/10.1177/0272989X08315241

Garrison LP, Pauly MV, Willke RJ, Neumann PJ. An overview of value, perspective, and decision context—a health economics approach: An ISPOR Special Task Force Report [2]. Value Health. 2018; 21:(2)124-130 https://doi.org/10.1016/j.jval.2017.12.006

Beckert S, Witte M, Wicke C A new wound-based severity score for diabetic foot ulcers: A prospective analysis of 1,000 patients. Diabetes Care. 2006; 29:(5)988-992 https://doi.org/10.2337/dc05-2431

Carter MJ, Gilligan AM, Waycaster CR Cost effectiveness of adding clostridial collagenase ointment to selective debridement in individuals with stage IV pressure ulcers. J Med Econ. 2017; 20:(3)253-265 https://doi.org/10.1080/13696998.2016.1252381

National Center for Health Statistics, Centers for Disease Control and Prevention. National Vital Statistics System: mortality data. 2013. https://tinyurl.com/37f6yf (accessed 19 June 2019)

Lázaro-Martínez JL, Aragón-Sánchez J, García-Morales E. Antibiotics versus conservative surgery for treating diabetic foot osteomyelitis: a randomized comparative trial. Diabetes Care. 2014; 37:(3)789-795 https://doi.org/10.2337/dc13-1526

Carter MJ, Fife CE. Clinic visit frequency in wound care matters: data from the US wound registry. J Wound Care. 2017; 26:S4-S10

Wilcox JR, Carter MJ, Covington S. Frequency of debridements and time to heal: a retrospective cohort study of 312 744 wounds. JAMA Dermatol. 2013; 149:(9)1050-1058 https://doi.org/10.1001/jamadermatol.2013.4960

The Centers for Medicare and Medicaid. Physician fee schedule search. https://tinyurl.com/hy2sdmq (accessed 19 June 2019)

The Centers for Medicare and Medicaid Services. Hospital Outpatient PPS. 2017. https://tinyurl.com/gwm6mmv (accessed 19 June 2019)

The Centers for Medicare and Medicaid Services. Durable medical equipment prosthetics/orthotics and supplies fee schedule. DME17-C. 2017. https://tinyurl.com/y4s7vj2e (accessed 19 June 2019)

The Centers for Medicare and Medicaid Services. 2017 Clinical diagnostic laboratory fee schedule. https://tinyurl.com/yy2ppdcrl (accessed 19 June 2019)

Centers for Medicare and Medicaid. Inpatient charge data, FY2015. https://tinyurl.com/y4km8fr9 (accessed 19 June 2019)

Redekop WK, Stolk EA, Kok E Diabetic foot ulcers and amputations: estimates of health utility for use in cost-effectiveness analyses of new treatments. Diabetes Metab. 2004; 30:(6)549-556 https://doi.org/10.1016/S1262-3636(07)70154-4

Sheehan P, Jones P, Giurini JM Percent change in wound area of diabetic foot ulcers over a 4-week period is a robust predictor of complete healing in a 12-week prospective trial. Plast Reconstr Surg. 2006; 117:239S-244S https://doi.org/10.1097/01.prs.0000222891.74489.33

Coerper S, Beckert S, Küper MA Fifty percent area reduction after 4 weeks of treatment is a reliable indicator for healing—analysis of a single-center cohort of 704 diabetic patients. J Diabetes Complications. 2009; 23:(1)49-53 https://doi.org/10.1016/j.jdiacomp.2008.02.001

Phillips TJ, Machado F, Trout R Prognostic indicators in venous ulcers. J Am Acad Dermatol. 2000; 43:(4)627-630 https://doi.org/10.1067/mjd.2000.107496

Alavi A, Sibbald RG, Phillips TJ What's new: Management of venous leg ulcers. J Am Acad Dermatol. 2016; 74:(4)643-664 https://doi.org/10.1016/j.jaad.2015.03.059

Malone M, Bjarnsholt T, McBain AJ The prevalence of biofilms in chronic wounds: a systematic review and meta-analysis of published data. J Wound Care. 2017; 26:(1)20-25 https://doi.org/10.12968/jowc.2017.26.1.20

Neumann PJ, Cohen JT, Weinstein MC. Updating cost-effectiveness—the curious resilience of the $50,000-per-QALY threshold. N Engl J Med. 2014; 371:(9)796-797 https://doi.org/10.1056/NEJMp1405158

Carter MJ. Health economics Information in wound care: the elephant in the room. Adv Wound Care. 2013; 2:(10)563-570 https://doi.org/10.1089/wound.2013.0479

Carter MJ. Cost-effectiveness research in wound care: definitions, approaches, and limitations. Ostomy Wound Manage. 2010; 56:(11)48-59

Siebert U, Alagoz O, Bayoumi AM State-Transition Modeling: A report of the ISPOR-SMDM Modeling Good Research Practices Task Force-3. Value Health. 2012; 15:(6)812-820 https://doi.org/10.1016/j.jval.2012.06.014

Eddy DM, Hollingworth W, Caro JJ Model transparency and validation: a report of the ISPOR-SMDM Modeling Good Research Practices Task Force-7. Value Health. 2012; 15:(6)843-850 https://doi.org/10.1016/j.jval.2012.04.012

Carter MJ, Fife CE, Walker D, Thomson B. Estimating the applicability of wound care randomized controlled trials to general wound-care populations by estimating the percentage of individuals excluded from a typical wound-care population in such trials. Adv Skin Wound Care. 2009; 22:(7)316-324 https://doi.org/10.1097/01.ASW.0000305486.06358.e0

Cost-utility of a biofilm-disrupting gel versus standard of care in chronic wounds: a Markov microsimulation model based on a randomised controlled trial

02 September 2022
Volume 6 · Issue 3

Abstract

Objective:

Analyse the cost-effectiveness and treatment outcomes of debridement (standard of care) plus BlastX, a biofilm-disrupting wound gel (group 1) or a triple-antibiotic, maximum-strength ointment (group 2), comparing a subset of patients who had not healed at four weeks using the ointment crossed-over to the biofilm-disrupting gel (group 3).

Methods:

A series of Markov microsimulation models were built using health states of an unhealed non-infected ulcer, healed ulcer, and infected non-healed ulcer and absorbing states of dead or amputation. All patients started with unhealed non-infected ulcers at cycle 0. Complications and healing rates were based on a randomised controlled trial (RCT). Costs were incurred by patients for procedures at outpatient wound care clinics and hospitals (if complications occurred) and were in the form of Medicare allowable charges. Quality-adjusted life years (QALYs) were computed using literature utility values. Incremental cost-effectiveness ratios (ICERs) were calculated for group 1 versus group 2, and group 3 versus group 2. One-way, multi-way and probabilistic sensitivity analysis (PSA) was conducted.

Results:

After one year, the base case ICER was $8794 per QALY for group 1 versus group 2, and $21,566 per QALY for group 3 versus group 2. Product cost and amputation rates had the most influence in one-way sensitivity analysis. PSA showed that the majority of costs were higher for group 1 but effectiveness values were always higher than for group 2. Average product use of 3.1ml per application represented 9.4% of the total group 1 cost (average $24.52 per application/$822.50 per group 1 patient). The biofilm-disrupting gel group performed substantially better than the current cost-effectiveness benchmarks, $8794 versus $50,000, respectively. Furthermore, when biofilm-disrupting gel treatment was delayed, as in group 3, the ICER outcomes were less substantial but it did remain cost-effective, suggesting the added benefits of immediate use of biofilm-disrupting gel. Also, when product cost assumptions used in the study were halved (Wolcott study usage), the model indicates important reductions in ICER to $966/QALY when comparing group 1 with group 2. It should be noted that product cost can hypothetically be affected not only by direct product purchase costs, but also by application intervals and technique. This suggests additional opportunities exist to optimise these parameters, maximising wound healing efficacy while providing significant cost savings to the payer.

Conclusion:

The addition of the biofilm-disrupting gel treatment to standard of care is likely to be cost-effective in the treatment of chronic wounds but when delayed by as little as 9–12 weeks the ICER is still far less than current cost-effectiveness benchmarks. The implication for payers and decision-makers is that biofilm-disrupting gel should be used as a first-line therapy at the first clinic visit rather than waiting as it substantially decreases cost-utility.

Acute wounds progress through several phases to final healing. When factors interfere with this orderly progression, this causes healing to stop and the wound to become chronic,1 and one of the most common factors for this is biofilm. Unlike planktonic bacteria, which exists as independent cells, biofilm is composed of microorganism aggregates protected by a non-crystalline extracellular matrix (ECM), extracellular polymeric substances (EPS), which is made of proteins, polysaccharides, lipids and other macromolecules.2,3,4,5

Biofilms frequently present without overt clinical symptoms of infection, are difficult to visualise except through imaging techniques due to their depth, and hard to eradicate by antibiotics because the tough EPS encapsulation makes penetration difficult. Moreover, with time, biofilm can adapt and evolve with cooperation of different species (quorum sensing), which provides further protection of bacteria from antibiotics and host defences. That said, the most important characteristics in wound healing impediment are the biofilm's virulence and pathogenicity,6 which may be dependent on the prevalence of less common species and their ability to form polymicrobial colonies.7,8,9

Register now to continue reading

Thank you for visiting Wound Central and reading some of our peer-reviewed resources for wound care professionals. To read more, please register today. You’ll enjoy the following great benefits:

What's included

  • Access to clinical or professional articles

  • New content and clinical updates each month