• Drug Coverage
  • Hypertrophic Cardiomyopathy (HCM)
  • Vaccines: 2023 Year in Review
  • Eyecare
  • Urothelial Carcinoma
  • Women's Health
  • Hemophilia
  • Heart Failure
  • Vaccines
  • Neonatal Care
  • NSCLC
  • Type II Inflammation
  • Substance Use Disorder
  • Gene Therapy
  • Lung Cancer
  • Spinal Muscular Atrophy
  • HIV
  • Post-Acute Care
  • Liver Disease
  • Pulmonary Arterial Hypertension
  • Safety & Recalls
  • Biologics
  • Asthma
  • Atrial Fibrillation
  • Type I Diabetes
  • RSV
  • COVID-19
  • Cardiovascular Diseases
  • Breast Cancer
  • Prescription Digital Therapeutics
  • Reproductive Health
  • The Improving Patient Access Podcast
  • Blood Cancer
  • Ulcerative Colitis
  • Respiratory Conditions
  • Multiple Sclerosis
  • Digital Health
  • Population Health
  • Sleep Disorders
  • Biosimilars
  • Plaque Psoriasis
  • Leukemia and Lymphoma
  • Oncology
  • Pediatrics
  • Urology
  • Obstetrics-Gynecology & Women's Health
  • Opioids
  • Solid Tumors
  • Autoimmune Diseases
  • Dermatology
  • Diabetes
  • Mental Health

Focus On: Dalbavancin A novel long-acting lipoglycopeptide antibiotic

News
Article

Abstract

Dalbavancin (Pfizer) is a new lipoglycopeptide antibiotic in phase 3 trials for the treatment of resistant gram-positive pathogens including methicillin-resistant Staphylococcus aureus (MRSA) and vancomycin-resistant enterococci (VRE). This agent exerts its bactericidal activity by binding to the terminal D-alanyl-D-alanine moiety of peptidoglycan precursors, thus blocking enzymes involved in the final stages of peptidoglycan synthesis and cell wall formation. Peak concentrations of dalbavancin occur immediately following infusion and increase in proportion to the dose given. After infusion, there is a rapid decline in plasma concentrations due to distribution of the drug into bodily tissues and fluids (Vss=0.52 L/kg). This initial rapid decline in plasma concentration is followed by a slower terminal, log-linear elimination phase, in which dalbavancin exhibits an elimination half-life ranging from 149 to 198 hours. Current data suggest that dalbavancin is well-tolerated by patients with most adverse events being described as mild. Dalbavancin may offer advantages over other antibiotics used in the treatment of resistant gram-positive pathogens because of its excellent tolerability and potency. Due to the long elimination half-life of dalbavancin, once-weekly dosing may be an option. (Formulary. 2006;41:59–73.)

Over the past 30 years, methicillin-resistant Staphylococcus aureus (MRSA) has evolved into a significant pathogen among hospitalized and non-hospitalized patients in the United States and around the world. The growing prevalence of MRSA infections not only poses a significant health risk, but also represents a substantial economic burden. Researchers have found that patients infected with MRSA have a longer average hospital stay of 4.5 days and a longer average antibiotic length of stay of 3 days when compared to patients infected with a methicillin-sensitive Staphylococcus aureus (MSSA). This translated into a median cost of $16,575 per patient per hospitalization, or approximately $4,000 more per hospitalization than a patient with a MSSA infection (2004 dollars).1

Although MRSA has been found among certain populations in the community, such as dialysis patients and intravenous drug users, for a number of years, this pathogen has recently become more prevalent among the general population.5 CDC researchers analyzed MRSA surveillance data from 3 US locations in 2001 and 2002 and found that between 8% and 20% of all MRSA infections were community-acquired.6 One study that examined infectious etiologies of diabetic foot ulcers at an outpatient clinic determined that MRSA was responsible for approximately 30% of foot infections in evaluated patients.7 In another study, MRSA was isolated from 61 of 137 patients (44.5%) presenting with localized skin or soft-tissue infections in a Los Angeles emergency department.8

As a result of the growing incidence of MRSA infections, empiric use of vancomycin has been increasing.9 Although vancomycin is commonly regarded as a potent agent against gram-positive microbes, its use is not without shortcomings. First, vancomycin appears to kill less efficiently than other agents such as the beta-lactams.10,11 This characteristic has led some clinicians to become concerned about slower rates of response to therapy when vancomycin is used. Second, vancomycin is only available as an intravenous agent and is routinely administered multiple times daily. Furthermore, most clinicians continue to monitor vancomycin serum concentrations and adjust therapy based on these levels. Additionally, vancomycin administration has been associated with several adverse events such as phlebitis and Red-man syndrome, which can complicate therapy. Finally, increased use of vancomycin has been associated with the emergence of resistant strains of Enterococcus species (VRE) and, more seriously, Staphylococcus aureus (VISA and VRSA). Vancomycin resistance among staphylococci has long been a concern, owing not only to the prevalence of the bacteria but also to the relatively few options that would be left to clinicians to combat this pathogen.2 Despite these problems, vancomycin remains the cornerstone of therapy for MRSA.

CHEMISTRY AND PHARMACOLOGY

Dalbavancin is a lipoglycopeptide antibiotic that is a semisynthetic derivative of the naturally occuring teicoplanin-like compound A40926. Similar to A40926 and teicoplanin, dalbavancin has a long fatty acyl moiety in an amide linkage to a glucosamine component. This fatty acyl chain is purported to improve activity by anchoring the molecule to the bacterial cell membrane, which prolongs the interaction of the agent with the bacteria.12

It has been postulated that dalbavancin exerts its bactericidal effect on bacteria through more than 1 mechanism. In addition to blocking enzymes involved in the final stages of peptidoglycan synthesis, dalbavancin may have a secondary mechanism of action independent of peptide binding. Researchers suggest that dalbavancin may inhibit transglycosylases, such as S aureus penicillin-binding protein 2 (PBP2), by direct interaction with enzymes involved in the final stages of peptidoglycan synthesis.14

Pharmacokinetics

Perhaps the most remarkable pharmacokinetic characteristic of dalbavancin is its long terminal half-life.Following single and multiple doses, the terminal half-life of dalbavancin has been reported to be 149 to 188 hours and 184 to 198 hours, respectively. It has been hypothesized that the long half-life of dalbavancin is likely due to the extensive protein-binding of the agent.17

Animal models have demonstrated that dalbavancin penetrates well into a variety of bodily tissues and fluids.20,21 In a study conducted by Jabes et al,21 granuloma pouches were induced in rats and then inoculated with 106 cfu/mL of MSSA and MRSA strains. Following a single dose of dalbavancin, it was noted that the drug demonstrated excellent penetration into the granuloma pouch. The ratio of plasma AUC to exudate AUC was approximately 1:1.21 Cavaleri et al20 examined the distribution of dalbavancin in 40 types of tissues. The investigators reported that following a single IV bolus dose, the highest concentrations of dalbavancin were found in the kidney and liver. Other tissues with measurable concentrations of dalbavancin included the heart, lung, spleen, and skin. The researchers observed that almost all tissues reached their maximum observed concentration within 24 hours of dose administration.

SPECTRUM OF ACTIVITY

Animal Models

Many animal models have been utilized to assess the efficacy of dalbavancin against various gram-positive pathogens. Candiani et al27 conducted a battery of experiments to examine the comparative efficacy of dalbavancin versus other antibiotics for the treatment of select gram-positive infections. In one arm of the study, the researchers compared dalbavancin, vancomycin, and teicoplanin for prophylaxis against septicemia in immunocompetent and neutropenic mice. The immunocompetent mice were infected with either 1.8x106 cfu of MSSA or 2.6x102 cfu of Streptococcus pneumoniae. Neutropenic mice were infected with either 1.1x105 cfu of Staphylococcus epidermidis or 3.2x104 cfu Enterococcus faecalis. Antibiotics were administered subcutaneously within 10 minutes following inoculation. A second dose of vancomycin was administered 5 hours after infection in those mice inoculated with S pneumoniae and S epidermidis. Efficacy was measured in terms of the dose that effectively protected 50% of the mice against septicemia (ED50). Results from this portion of the study indicated that dalbavancin and teicoplanin possess similar protective potency, whereas vancomycin was much less active against MSSA (ED50=0.08, 0.2, and 1.12 mg/kg/dose, respectively). The activities of teicoplanin and dalbavancin against S pneumoniae were slightly better than the double-dose of vancomycin (ED50=0.56, 0.40, and 0.79 mg/kg/dose, respectively). Against E faecalis, teicoplanin was more effective than dalbavancin in preventing septicemia in infected mice (0.51 and 1.53 mg/kg/dose, respectively). Vancomycin was not tested against E faecalis.

Lefort et al30 also examined the efficacy of dalbavancin in an animal model of endocarditis. In this study, researchers inoculated rabbits with 1 of 3 strains of S aureus with reduced susceptibility to vancomycin and teicoplanin. Following inoculation, animals were administered either a single 40 mg/kg IV dose of dalbavancin or a 10 mg/kg daily IV dose for 4 days. Susceptibility testing revealed that dalbavancin was 2 to 4 times more potent than vancomycin and 4 to 8 times more potent than teicoplanin against these isolates. Data of bacterial density in vegetations demonstrated that the multiple-dose dalbavancin regimen decreased bacterial counts by 3.5 to 3.9 log10 cfu/g of vegetation, while the single-dose dalbavancin regimen reduced bacterial counts by 2.1 to 2.5 cfu/g of vegetation (P<.01).

CLINICAL TRIALS

Data from a clinical trial regarding the use of dalbavancin for complicated skin and skin structure infections (cSSSIs) were recently been published in abstract form.32 This was a randomized (2:1, dalbavancin:linezolid), double-blind, comparative trial of dalbavancin (1,000 mg IV on Day 1 and 1,500 mg on Day 8) versus linezolid (600 mg IV every 12 or IV followed by oral therapy in a sequential manner) for the treatment of cSSSIs. Clinical and microbiologic efficacy were each evaluated at the test-of-cure visit. A total of 434 and 226 patients were available for clinical evaluation in the dalbavancin and linezolid groups, respectively. Two hundred seventy-seven dalbavancin-treated and 152 linezolid-treated patients were microbiologically evaluable. The primary skin and skin structure infections (SSSIs) noted among the study population included major abscess (32.3%) and cellulitis (28.2%). Staphylococcus aureus was the most common pathogen identified. Fifty-six percent of the S aureus isolates were resistant to methicillin. Success rates (clinical/microbiologic) of 88.9%/89.5% and 91.2%/ 87.5% were reported for dalbavancin and linezolid, respectively. The investigators noted that <1% of patients in either treatment group experienced a relapse. These findings led the authors to conclude that 2 doses of dalbavancin administered 1 week apart were equally as effective as a twice-daily dosing regimen of linezolid for 14 days.

Data from three phase 3 comparative studies of dalbavancin versus either linezolid, cefazolin, or vancomycin for the treatment of skin and SSSIs were also recently published in abstract form.33 Dalbavancin was dosed once-weekly and comparators were administered via conventional dosing regimens. Responses were evaluated using clinical and microbiologic end points. Seventy-seven percent of the pathogens recovered were S aureus, and 38% of staphylococcal isolates were methicillin- resistant. Among the 3 studies, the clinical response rate for dalbavancin ranged from 89% to 90% and was similar to the response rates noted for comparator agents. Similarly, microbiologic eradication rates were similar among the study medications. These data led the investigators to conclude that dalbavancin was as equally effective as comparator agents for the treatment of skin and SSSIs caused by S aureus.

The efficacy of dalbavancin for the treatment of catheter-related bloodstream infections caused by gram-positive pathogens has also been studied. In an open-label trial, 75 adult patients with catheter-related bloodstream infections (CR-BSIs) suspected to be caused by coagulase-negative staphylococci or S aureus, including MRSA, were randomized to receive either a multiple-dose dalbavancin regimen consisting of a single 1,000-mg IV dose followed by a single 500-mg IV dose 1 week later, or IV vancomyin administered twice daily for 14 days. Twenty-three patients in the dalbavancin group and 28 patients in the vancomycin group were evaluable for the primary efficacy end point of clinical success at the test-of-cure visit. Overall success rates for the primary efficacy end point revealed that dalbavancin appeared to be superior to vancomycin for the treatment of CR-BSIs (87%; 95% CI, 73.2%–100% vs 50%; 95% CI, 31.5%–68.5%).32

Adverse Events

Current data demonstrate that dalbavancin is well-tolerated by patients, with most adverse events being described as mild.15,30–32 In a proof-of-concept study assessing the efficacy and safety of dalbavancin for SSTIs using an intravenous 2-dose scheme of 1,000 mg followed by a 500-mg dose 1 week later or a 1-time 1,100-mg dose, no serious adverse events were reported. Additionally, no discontinuations of therapy were reported that resulted from adverse events due to dalbavancin use. Also, no clinically relevant laboratory value alterations, including serum creatinine elevations, were observed.31 Another study using a similar dosing strategy determined that dalbavancin use was not associated with any serious adverse events in study subjects. In this study, the most commonly reported adverse events believed to be related to dalbavancin use included oral candidiasis, diarrhea, constipation, and febrile response. No patients in the dalbavancin group of this study discontinued treatment due to adverse events. Another trial, utilizing intravenously administered multiple-dose and escalating single-dose regimens of dalbavancin, discovered that dalbavancin was well-tolerated and no dose-limiting toxicities were encountered. The most commonly reported adverse events were pyrexia, headache, and nausea.15 The potential for ototoxicity with dalbavancin use was also determined to be low in phase 1 trials.33

Safety data from the intent-to-treat (ITT) populations for the phase 2/3 development trials of dalbavancin were recently summarized.36 In these studies a total of 1,126 patients received at least 1 dose of dalbavancin. It was noted that 52% of subjects who had received dalbavancin reported at least 1 adverse event. This is comparable to the 56.9% of subjects enrolled in the comparator arms of these studies. Of these events, 3.5% and 3.8% resulted in discontinuation of the study medication in the dalbavancin and comparator groups, respectively.

Of note, histamine-related adverse events such as flushing, hypotension, and rash reactions that have been described for other glycopeptides have not been reported following dalbavancin administration.

Although dalbavancin may not appear to pose a significant risk for adverse events, consideration should be given to how this agent's pharmacokinetic profile impacts the overall clinical picture. Due to dalbavancin's long half-life, reversal of an adverse event could take an extended period of time.

DOSING AND ADMINISTRATION

Several dosing regimens have been used with dalbavancin in clinical trials. Both one-time doses ranging from 140 mg to 1,120 mg, and multiple-dose regimens with loading doses ranging from 300 mg to 1,000 mg and maintenance doses ranging from 30 mg to 500 mg, have been utilized.15,31,32 The one-time IV bolus dose of 1,120 mg is the highest dose administered to human subjects, and it was well tolerated.15,16 Although no dosing regimen for dalbavancin has been approved, it appears that a multiple-dose regimen of 1,000 mg followed by a 500-mg dose 1 week later may be the most likely regimen. This dosing regimen was determined to be safe and effective for the treatment of infections caused by S aureus and coagulase-negative staphylococci-related infections. The administration of dalbavancin was via IV infusion over the course of 30 minutes in clinical trials.15,31

Drug Interactions

Mr Blostica is a PharmD candidate at Ferris State University, Kalamazoo, Mich. He can be reached at tblostica@yahoo.com
.

Dr Klepser is a pharmacy practice professor at Ferris State University.

In each issue, the "Focus on" feature reviews a newly approved or investigational drug of interest to pharmacy and therapeutics committee members. The column is coordinated by Robert A. Quercia, MS, RPh, director of Drug Information Services at Hartford Hospital in Hartford, Conn, and adjunct associate professor, University of Connecticut School of Pharmacy, Storrs, Conn; and by Craig I. Coleman, PharmD, assistant professor of pharmacy practice, University of Connecticut School of Pharmacy, and director, Pharmacoeconomics and Outcomes Studies Group, Hartford Hospital.

Editors' note: The clinical information provided in "Focus on" articles is as current as possible. Due to regularly emerging data on developmental or newly approved drug therapies, articles include information published or presented and available to the author up until the time of the manuscript submission.

REFERENCES

1. Kopp BJ, Nix DE, Armstrong EP. Clinical and economic analysis of methicillin-susceptible and -resistant Staphylococcus aureus infections. Ann Pharmacother. 2004;38:1377–1382.

2. Kuehnert MJ, Hill HA, Kupronis BA, Tokars JI, Solomon SL, Jernigan DB. Methicillin-resistant-Staphylococcus aureus hospitalizations, United States. Emerg Infect Dis. 2005;11:868–872.

3. Baquero F. Gram-positive resistance: challenge for the development of new antibiotics. J Antimicrob Chemother. 1997;39(Suppl A):1–6.

4. Excerpt from annual communicable disease report 2002. Washington State Department of Health. Appendix 4. 106-9. Available at: http:// http://www.doh.wa.gov/Topics/Antibiotics/appIV.pdf . Accessed January 31, 2006.

5. Arnold FW, Wojda B. An analysis of a community-acquired pathogen in a Kentucky community: methicillin-resistant Staphylococcus aureus. J Ky Med Assoc. 2005;103:206–210.

6. Fridkin SK, Hageman JC, Morrison M, et al. Methicillin-resistant Staphylococcus aureus disease in three communities. N Engl J Med. 2005;352:1436–1444.

7. Dang CN, Prasad YD, Boulton AJ, Jude EB. Methicillin-resistant Staphylococcus aureus in the diabetic foot clinic: a worsening problem. Diabet Med. 2003;20:159-161.

8. Frazee BW, Lynn J, Charlebois ED, Lambert L, Lowery D, Perdreau-Remington F. High prevalence of methicillin-resistant Staphylococcus aureus in emergency department skin and soft tissue infections. Ann Emerg Med. 2005;45(3):311–320.

9. Hamilton-Miller JM. Vancomycin-resistant Staphylococcus aureus: a real and present danger? Infection. 2002;30:118–124.

10. Small PM, Chambers HF. Vancomycin for Staphylococcus aureus endocarditis in intravenous drug users. Antimicrob Agents Chemother. 1990;34:1227–1231.

11. Cantoni L, Glauser MP, Bille J. Comparative efficacy of daptomycin, vancomycin, and cloxacillin for the treatment of Staphylococcus aureus endocarditis in rats and role of test conditions in this determination. Antimicrob Agents Chemother. 1990;34:2348–2353.

12. Beauregard DA, Williams DH, Gwynn MN, Knowles DJ. Dimerization and membrane anchors in extracellular targeting of vancomycin group antibiotics. Antimicrob Agents Chemother. 1995;39:781–785.

13. Perkins HR. Specificity of combination between mucopeptide precursors and vancomycin or ristocetin. Biochem J. 1969;111:195–205.

14. Leimkuhler C, Chen L, Barrett D, et al. Differential inhibition of Staphylococcus aureus PBP2 by glycopeptide antibiotics. J Am Chem Soc. 2005;127:3250–3251.

15. Leighton A, Gottlieb AB, Dorr MB, et al. Tolerability, pharmacokinetics, and serum bactericidal activity of intravenous dalbavancin in healthy volunteers. Antimicrob Agents Chemother. 2004;48:940–945.

16. Leighton A, Mroszcak E, White R, et al. Dalbavancin: phase 1 single and multiple-dose placebo controlled intravenous safety pharmacokinetic study in healthy volunteers. Presented at: 41st Interscience Conference of Antimicrobial Agents and Chemotherapy; 2001; Chicago, Ill. Abstract 951.

17. Cavaleri M, Cooper A, Nutley MA, Stognew M. Protein binding of dalbavancin using isothermal titrationo microcalorimetry. Presented at: 42nd Interscience Conference on Antimicrobial Agents and Chemotherapy; 2002; San Diego, Calif. Abstract A-1385.

18. Matzke GR, Zhanel GG, Guay DR. Clinical pharmacokinetics of vancomycin. Clin Pharmacokinet. 1986;11:257–282.

19. Blouin RA, Bauer LA, Miller DD, Record KE, Griffen WO, Jr. Vancomycin pharmacokinetics in normal and morbidly obese subjects. Antimicrob Agents Chemother. 1982;21:575–580.

20. Cavaleri M, Riva S, Valagussa A, et al. Pharmacokinetics and excretion of dalbavancin in the rat. J Antimicrob Chemother. 2005;55(Suppl 2):31–35.

21. Jabes D, Candiani G, Romano G, Brunati C, Riva S, Cavaleri M. Efficacy of dalbavancin against methicillin-resistant Staphylococcus aureus in the rat granuloma pouch infection model. Antimicrob Agents Chemother. 2004;48: 1118–1123.

22. Streit JM, Fritsche TR, Sader HS, Jones RN. Worldwide assessment of dalbavancin activity and spectrum against over 6,000 clinical isolates. DiagnMicrobiol Infect Dis. 2004;48:137–143.

23. Mushtaq S, Warner M, Johnson AP, Livermore DM. Activity of dalbavancin against staphylococci and streptococci, assessed by BSAC and NCCLS agar dilution methods. J Antimicrob Chemother. 2004;54:617–620.

24. Gales AC, Sader HS, Jones RN. Antimicrobial activity of dalbavancin tested against Gram-positive clinical isolates from Latin American medical centres. Clin Microbiol Infect. 2005;11:95–100.

25. Lopez S, Hackbarth C, Romano G, Trias J, Jabes D, Goldstein BP. In vitro antistaphylococcal activity of dalbavancin, a novel glycopeptide. J Antimicrob Chemother. 2005;55(Suppl 2):21–24.

26. Jones RN, Biedenbach DJ, Johnson DM, Pfaller MA. In vitro evaluation of BI 397, a novel glycopeptide antimicrobial agent. J Chemother. 2001;13:244–254.

27. Candiani G, Abbondi M, Borgonovi M, Romano G, Parenti F. In-vitro and in-vivo antibacterial activity of BI 397, a new semi-synthetic glycopeptide antibiotic. J Antimicrob Chemother. 1999;44:179–192.

28. Goldstein EJ, Citron DM, Merriam CV, Warren Y, Tyrrell K, Fernandez HT. In vitro activities of dalbavancin and nine comparator agents against anaerobic gram-positive species and corynebacteria. Antimicrob Agents Chemother. 2003;47: 1968–1971.

29. Harding I, MacGowan AP, White LO, Darley ES, Reed V. Teicoplanin therapy for Staphylococcus aureus septicaemia: relationship between pre-dose serum concentrations and outcome. J Antimicrob Chemother. 2000;45:835–841.

30. Lefort A, Pavie J, Garry L, Chau F, Fantin B. Activities of dalbavancin in vitro and in a rabbit model of experimental endocarditis due to Staphylococcus aureus with or without reduced susceptibility to vancomycin and teicoplanin. Antimicrob Agents Chemother. 2004;48:1061–1064.

31. Seltzer E, Dorr MB, Goldstein BP, Perry M, Dowell JA, Henkel T. Once-weekly dalbavancin versus standard-of-care antimicrobial regimens for treatment of skin and soft-tissue infections. Clin Infect Dis. 2003;37:1298–1303.

32. Jauregui-Paredo L ORW, Goldberg L, Wible M, Seltzer E. Efficacy and safety of weekly dalbavancin versus linezolid in complicated skin and skin structure infections. Presented at: 45th Interscience Conference of Antimicrobial Agents and Chemotherapy; 2005; Washington, DC. Abstract L-1575.

33. Goldstein B, Seltzer E, Flamm R, Sahm D. Dalbavancin phase 3 skin and skin structure (SSSI) studies: pathogens and microbiological efficacy. Presented at: 45th Interscience Conference of Antimicrobial Agents and Chemotherapy; 2005; Washington, DC. Abstract L-1577.

34. Raad I, Darouiche R, Vazquez J, et al. Efficacy and safety of weekly dalbavancin therapy for catheter-related bloodstream infection caused by gram-positive pathogens. Clin Infect Dis. 2005;40:374–380.

35. Campbell KC, Kelly E, Targovnik N, et al. Audiologic monitoring for potential ototoxicity in a phase I clinical trial of a new glycopeptide antibiotic. J Am Acad Audiol. 2003;14:157–168.

36. Seltzer E, Goldberg L, Krause D. Safety of dalbavancin in a clinical development program. Presented at: 45th Interscience Conference of Antimicrobial Agents and Chemotherapy; 2005; Washington, DC. Abstract L-1576.

37. Drug Facts and Comparisons. 2004. St Louis, Mo: Facts and Comparisons, Inc.

Related Videos
Related Content
© 2024 MJH Life Sciences

All rights reserved.