Showing posts sorted by date for query Vancomycin. Sort by relevance Show all posts
Showing posts sorted by date for query Vancomycin. Sort by relevance Show all posts

Thursday, April 8, 2021

FDA Approves Kimyrsa (oritavancin) for the Treatment of Adult Patients with Acute Bacterial Skin and Skin Structure Infections (ABSSSI)


Melinta Therapeutics, LLC (Melinta), a commercial-stage company focused on the development and commercialization of novel antibiotics, today announced that the U.S. Food and Drug Administration (FDA) has approved Kimyrsa (oritavancin)  for the treatment of adult patients with acute bacterial skin and skin structure infections (ABSSSI) caused by susceptible isolates of designated Gram-positive microorganisms, including methicillin-resistant Staphylococcus aureus (MRSA). Kimyrsa is a lipoglycopeptide antibiotic that delivers a complete course of therapy for ABSSSI in a single, one hour 1,200 mg infusion.



“The approval of Kimyrsa demonstrates Melinta’s commitment to provide innovative therapies to patients with acute and life-threatening illnesses,” said Christine Ann Miller, President and Chief Executive Officer of Melinta. “We have responded to the requests of the medical community to provide an oritavancin product with a shorter infusion time.  We believe that with the approval of Kimyrsa and product availability this summer, physicians and patients will now have a compelling new one-dose alternative to the current standard of multi-dose regimens for ABSSSI.”

ABSSSI affect approximately 14 million patients in the U.S. each year, are responsible for over 3 million visits to the Emergency Room annually and represent the 8th most common cause of Emergency Department hospital admissions1,2. ABSSSI cost U.S. hospitals $4 billion each year, with a 4.1-day average length of stay for hospitalized ABSSSI patients.2

“Kimyrsa is an important new treatment option that will provide clinicians with additional flexibility to treat ABSSSI patients in multiple care settings, without the need for hospitalization,” said Andrew Dold, D.O., member of a private infectious disease practice covering the Greater Atlanta Region. “Single-dose, long-acting antibiotics, such as Kimyrsa, may be especially beneficial for patients who lack the support or resources to adhere to multiple intravenous administrations.”

The efficacy and safety of Kimyrsa were established in the SOLO clinical trials with another oritavancin product, Orbactiv. The SOLO trials were randomized, double-blind, multicenter studies that evaluated a single 1,200 mg IV dose of oritavancin against twice-daily vancomycin for the treatment of ABSSSI in 1,987 adult patients and assessed one of the largest subsets of documented MRSA infection (405 patients). These trials demonstrated that 1,200 mg one-dose IV oritavancin infusion was as effective as 7-to-10 days of twice-daily vancomycin (1 g or 15 mg/kg) for the primary and secondary endpoints.  Kimyrsa approval is based on the results of an open-label, multi-center, pharmacokinetics study, which compared Kimyrsa administered over 1 hour (N=50) to Orbactiv administered over 3 hours (N=52) for the treatment of adult patients with ABSSSI.

Michael Waters, M.D. and lead investigator in the PK clinical trial stated, “Kimyrsa was shown to be comparable to Orbactiv with a favorable safety profile.  I’m pleased that these outcomes support the approval of Kimyrsa to provide oritavancin with a shorter infusion time and lower infusion volume.  With these features, Kimyrsa can further enhance the treatment experience for the patient and efficiency of administration in clinical practice.”

https://en.wikipedia.org/wiki/Oritavancin

Friday, April 20, 2018

CutisPharma Announces FDA Approval of Firvanq (vancomycin) for Treatment of Clostridium Difficile Associated Diarrhea and Staphylococcus Aureus Colitis


In continuation of my update on vancomycin

Vancomycin.svg

CutisPharma announced  the US Food and Drug Administration (FDA)  approval of  Firvanq (vancomycin hydrochloride) for oral solution, for the treatment of Clostridium difficile associated diarrhea and enterocolitis caused by Staphylococcus aureus, including methicillin-resistant strains.

“We are pleased to announce the FDA approval of Firvanq,” said Neal I. Muni, MD, MSPH, Chief Executive Officer of CutisPharma. “Firvanq's approval is an important step forward to providing patients the only FDA-approved vancomycin oral liquid treatment option for Clostridium difficile associated diarrhea, a life-threatening condition that affects over a half-million patients in the United States annually.”
Upon its launch, which is targeted to be April 2, 2018, Firvanq will replace CutisPharma’s FIRST®-Vancomycin Unit-of-Use Compounding Kit, which has been available to pharmacists that need a convenient, accurate, and compliant way to compound vancomycin oral liquid therapy. Firvanq will be commercially available in 25 mg/mL and 50 mg/mL strengths in convenient 150 mL and 300 mL sizes. Firvanq is designed to be easy to use and has the potential to be a cost-effective alternative to existing vancomycin therapies.
“As a practicing infectious disease physician treating many patients with CDAD, having an FDA-approved vancomycin oral liquid formulation that is affordable and accessible to my patients is very beneficial,” said Stuart Johnson, MD, Loyola University Medical Center. “Patient access is currently limited by the fact that only a select few pharmacies perform compounding in the outpatient setting these days, given the many new regulations in place. Availability of an FDA-approved vancomycin oral liquid treatment will effectively allow any pharmacy to stock this therapy, and hopefully encourage third-party payer reimbursement, significantly improving accessibility and convenience for patients.”

Wednesday, February 7, 2018

CutisPharma Announces FDA Approval of Firvanq (vancomycin) for Treatment of Clostridium Difficile Associated Diarrhea and Staphylococcus Aureus Colitis

In continuation of my update on Firvanq
Vancomycin.svg
CutisPharma announced that the US Food and Drug Administration (FDA) has approved Firvanq (vancomycin hydrochloride) for oral solution, for the treatment of Clostridium difficile associated diarrhea and enterocolitis caused by Staphylococcus aureus, including methicillin-resistant strains.
“We are pleased to announce the FDA approval of Firvanq,” said Neal I. Muni, MD, MSPH, Chief Executive Officer of CutisPharma. “Firvanq's approval is an important step forward to providing patients the only FDA-approved vancomycin oral liquid treatment option for Clostridium difficile associated diarrhea, a life-threatening condition that affects over a half-million patients in the United States annually.”
Upon its launch, which is targeted to be April 2, 2018, Firvanq will replace CutisPharma’s FIRST®-Vancomycin Unit-of-Use Compounding Kit, which has been available to pharmacists that need a convenient, accurate, and compliant way to compound vancomycin oral liquid therapy. Firvanq will be commercially available in 25 mg/mL and 50 mg/mL strengths in convenient 150 mL and 300 mL sizes. Firvanq is designed to be easy to use and has the potential to be a cost-effective alternative to existing vancomycin therapies.
“As a practicing infectious disease physician treating many patients with CDAD, having an FDA-approved vancomycin oral liquid formulation that is affordable and accessible to my patients is very beneficial,” said Stuart Johnson, MD, Loyola University Medical Center. “Patient access is currently limited by the fact that only a select few pharmacies perform compounding in the outpatient setting these days, given the many new regulations in place. Availability of an FDA-approved vancomycin oral liquid treatment will effectively allow any pharmacy to stock this therapy, and hopefully encourage third-party payer reimbursement, significantly improving accessibility and convenience for patients.”
Ref : https://www.drugs.com/history/firvanq.html

Friday, December 30, 2016

Allergan Receives FDA Approval of Teflaro (ceftaroline fosamil) for Pediatric Patients

Ceftaroline fosamil2DCSD.svg

Allergan plc, a leading global pharmaceutical company,  announced the U.S. Food and Drug Administration (FDA) has approved the company's supplemental New Drug Application (sNDA) for Teflaro (ceftaroline fosamil), granting new indications for pediatric patients 2 months of age to less than 18 years of age with acute bacterial skin and skin structure infections (ABSSSI), including infections caused by methicillin-resistant Staphylococcus aureus (MRSA), and community-acquired bacterial pneumonia (CABP) caused by Streptococcus pneumoniae and other designated susceptible bacteria.

"The impact of ABSSSI and CABP among children is significant, as these infections often require hospitalization and are met with limited pediatric treatment options, particularly as resistance increases among the pathogens that cause these infections," said David Nicholson, Chief R&D Officer, Allergan. "These new indications are yet another testament to our ongoing research and development in anti-infectives to address the evolving challenges of serious infections. Importantly, it allows us to educate physicians on the data they need to prescribe Teflaro to appropriate pediatric patients in need of an option that is safe and effective against some of the most difficult-to-treat pathogens in ABSSSI and CABP."
ABSSSI and CABP are common causes of healthcare visits and hospitalizations among children. Studies show more than 70,000 hospitalizations for ABSSSI occur among children per year – a rate that has more than doubled over the past 13 years.1 A study conducted by the Centers for Disease Control and Prevention (CDC) also found children younger than 5 years of age accounted for 70 percent of children hospitalized for community-acquired pneumonia.
These new indications were approved based on results from clinical studies evaluating TEFLARO in pediatric patients (2 months to less than 18 years of age), including one active-controlled study in ABSSSI and two active-controlled studies in CABP. In the ABSSSI active-controlled study, the efficacy and safety of Teflaro was compared with vancomycin or cefazolin (each with optional aztreonam). In the CABP studies, Teflaro was compared with ceftriaxone. Use of Teflaro in pediatric patients 2 months to less than 18 years of age is supported by evidence from adequate and well-controlled studies of Teflaro in adults, as well as additional pharmacokinetic and safety data from pediatric trials.
The primary objective of the pediatric ABSSSI and CABP studies was to evaluate the safety and tolerability of Teflaro. These studies were not powered for comparative inferential efficacy analysis, and no efficacy endpoints were identified as primary.
To evaluate the treatment effect of Teflaro in the ABSSSI pediatric trial, an analysis was conducted in 159 patients with ABSSSI in the Modified Intent-to-Treat (MITT) population. This analysis evaluated responder rates based on achieving both cessation of lesion spread and absence of fever on Study Day 3. Patients treated with Teflaro showed a higher response at Study Day 3 versus the comparator group, with clinical response achieved in 80.4 percent (86/107) of patients treated with Teflaro and 75 percent (39/52) of patients in the comparator group, with a treatment difference of 5.4 percent (95 percent Confidence Interval [CI] -7.8, 20.3). Clinical cure rates at the test of cure (TOC) visit (8 to 15 days after the end of therapy) for the ABSSSI pediatric trial were 94.4 percent (101/107) for patients treated with Teflaro and 86.5 percent (45/52) for the comparator, with a treatment difference of 7.9 (95 percent CI -1.2, 20.2).
To evaluate the treatment effect of Teflaro in the CABP trial submitted for this pediatric filing, an analysis was conducted in 143 patients with CABP in the MITT population. This analysis evaluated responder rates at Study Day 4 based on achieving improvement in at least two out of seven symptoms (cough, dyspnea, chest pain, sputum production, chills, feeling of warmth/feverish and exercise intolerance or lethargy), and worsening in none of these symptoms. The clinical response at Study Day 4 was 69.2 percent (74/107) for patients treated with Teflaro and 66.7 percent (24/36) for the comparator, with a treatment difference of 2.5 percent (95 percent CI -13.9, 20.9). Clinical cure rates at TOC were 87.9 percent (94/107) for patients treated with TEFLARO and 88.9 percent (32/36) for the comparator, with a treatment difference of -1.0 (95 percent CI -11.5, 14.1).
Results from the clinical studies in pediatric patients showed that Teflaro demonstrated a safety profile that was compatible with treatment of ABSSSI and CABP at the clinical dosages studied. The safety findings were similar to those seen in the adult studies, and no safety concerns were identified beyond those already known to be cephalosporin class effects.
Teflaro is the first and only cephalosporin indicated in adults and pediatric patients 2 months of age and older for the treatment of ABSSSI and CABP due to designated susceptible pathogens that can be administered by intravenous (IV) infusion in five minutes to one hour.

Thursday, December 1, 2016

Theravance Biopharma Announces FDA Approval of Expanded Label for Vibativ (telavancin)

In continuation of my update on telavancin

Telavancin.png
Theravance Biopharma, Inc. (NASDAQ: TBPH) ("Theravance Biopharma" or the "Company") today announced that the U.S. Food and Drug Administration (FDA) has approved the Company's supplemental New Drug Application (sNDA) for Vibativ (telavancin) to expand the product's label to include data describing the treatment of patients with concurrent Staphylococcus aureus (S. aureus) bacteremia in both of the antibiotic's currently approved indications in the United States. Vibativ is approved in the U.S. for the treatment of adult patients with hospital-acquired and ventilator-associated bacterial pneumonia (HABP/VABP) caused by susceptible isolates of S. aureus when alternative treatments are not suitable. In addition, Vibativ is approved in the U.S. for the treatment of adult patients with complicated skin & skin structure infections (cSSSI) caused by susceptible isolates of Gram-positive bacteria, including S. aureus, both methicillin-susceptible (MSSA) and methicillin-resistant (MRSA) strains.
Bacteremia is the presence of bacteria in the bloodstream and can occur spontaneously or in the presence of other infections. Bacteremia continues to represent a significant unmet medical need. Concurrent bacteremia, which in its most serious form is fatal, occurs when bacteria spreads from its initial infection site and enters the bloodstream. As a secondary infection, it introduces significant challenges to the treatment of the primary infection, as well as the concurrent bacteremia itself.
"When patients with cSSSI or HABP/VABP present with concurrent bacteremia, their treatment becomes more difficult for healthcare practitioners. With this action by the FDA, we believe an important advance has been made in addressing the unique challenges in this area. The additional data that are now reflected in the Vibativ label address the use of Vibativ in cSSSI and HABP/VABP with concurrent bacteremia and we are now implementing a strategy to communicate this information to targeted healthcare practitioners," said Frank Pasqualone, Senior Vice President and Global Head, Acute Care Business at Theravance Biopharma. "With the broader medical need for effective bacteremia treatments in mind, we are also conducting a Phase 3 registrational study of telavancin in primary complicated S. aureus bacteremia, which we expect to complete in late 2017 or early 2018. Should we prove successful with this trial and secure approval in this infection type, Vibativ would possess the broadest set of indications of any branded anti-MRSA agent."
The sNDA filing was based on the combined data from Theravance Biopharma's previously conducted pivotal trials of Vibativ in its two approved indications -- cSSSI (ATLAS I and II) and HABP/VABP (ATTAIN I and II). The trials were large, multi-center, multinational, double-blind, randomized Phase 3 clinical studies enrolling and treating 3,370 adult patients, including a portion of patients with concurrent bacteremia. Importantly, these studies involved two of the largest cohorts of patients ever studied in these diseases and included one of the largest cohorts of patients with MRSA infections studied to date.

Expanded Vibativ Label Data

The data added to the Vibativ label describe patients with concurrent S. aureus bacteremia in the Phase 3 ATLAS and ATTAIN trials. These include:
  • In the all-treated cSSSI patient population with baseline S. aureus bacteremia in the ATLAS I and II trials, clinical cure rates at test-of-cure were 57.1% for Vibativ-treated patients vs. 54.6% for vancomycin-treated patients.
  • In the HABP/VABP patient population with at least one Gram-positive respiratory pathogen at baseline who had concurrent S. aureus bacteremia in the ATTAIN I and II trials, the 28-day all-cause mortality rate was 40.0% for Vibativ-treated patients vs. 39.5% for vancomycin-treated patients.
Separately, Theravance Biopharma is currently conducting a Phase 3 registrational study of telavancin in patients with complicated S. aureus bacteremia. The trial is a multi-center, randomized, open-label study that is enrolling approximately 250 adult patients with confirmed MSSA or MRSA bacteremia at about 70 clinical sites in the U.S. and around the world. Researchers are evaluating telavancin in treating these patients as compared to standard therapies such as vancomycin, daptomycin and anti-staphylococcal penicillins. The trial is expected to be completed in late 2017 or early 2018.

Friday, October 28, 2016

Natural product darwinolide may help combat fatal MRSA infection

A serious and sometimes fatal bacterial infection, known as methicillin-resistantStaphylococcus aureus (MRSA), may soon be beatable thanks to the efforts of University of South Florida scientists who have isolated and tested an extract from a sponge found in Antarctica. The sponge extract, known as Dendrilla membranosa, yields a new, natural product chemical which has shown in laboratory tests that it can eliminate more than 98 percent of MRSA cells. The research team has named the new chemical "darwinolide."

Image result for darwinolide


The study describing their methods and results was published this week in the American Chemical Society's journal Organic Letters.
While years ago the highly-resistant MRSA infection was particularly problematic in places such as hospitals and nursing homes, it has developed into an infection that can be found in commonly-used places such as gyms, locker rooms and schools.
"In recent years, MRSA has become resistant to vancomycin and threatens to take away our most valuable treatment option against staph infections," said study co-author and USF microbiologist Dr. Lindsey N. Shaw.
MRSA is unique in that it can cause infections in almost every niche of the human host, from skin infections, to pneumonia, to endocarditis, a serious infection of tissues lining the heart. Unfortunately, the pace of the pharmaceutical industry's efforts to find new antibiotics to replace those no longer effective has slowed in recent years, said Shaw.
Like many other bacterium, the MRSA bacteria forms a biofilm.
"Biofilms, formed by many pathogenic bacteria during infection, are a collection of cells coated in a variety of carbohydrates, proteins and DNA," said Shaw. "Up to 80 percent of all infections are caused by biofilms and are resistant to therapy. We desperately need new anti-biofilm agents to treat drug resistant bacterial infections like MRSA."
USF chemistry professor Dr. Bill Baker and colleagues have literally gone to the 'ends of the Earth' to help in the fight against MRSA. Baker, who also serves as director of the USF Center for Drug Discovery and Innovation (CDDI), studies the chemical ecology of Antarctica and dives in the frigid waters near Palmer Station to retrieve marine invertebrates, such as sponges, to carry out "natural product isolation," which means drawing out, modifying and testing natural substances that may have pharmaceutical potential.
His group led the effort to extract and characterize chemical structures to create darwinolide from the freeze-dried Antarctic sponges and then test in Shaw's lab to determine its effectiveness against the MRSA bacteria.

"When we screened darwinolide against MRSA we found that only 1.6 percent of the bacterium survived and grew. This suggests that darwinolide may be a good foundation for an urgently needed antibiotic effective against biofilms," said Baker, whose research team "rearranged" the chemical composition of the extracted sponge.

In the last 70 years, despite the discovery and use of antibiotics to treat infections, bacterial disease remains the second-leading cause of death globally, especially among children and the elderly, noted the researchers. In the U.S. alone there are two million hospital acquired infections annually with at least 100,000 deaths, many resulting from bacteria resistant to current antibiotics.
"We suggest that darwinolide may present a highly suitable scaffold for the development of urgently needed, novel, anti-biofilm-specific antibiotics," concluded the researchers.

Ref : http://pubs.acs.org/doi/abs/10.1021/acs.orglett.6b00979?journalCode=orlef7




Natural product darwinolide may help combat fatal MRSA infection: A serious and sometimes fatal bacterial infection, known as methicillin-resistant Staphylococcus aureus (MRSA), may soon be beatable thanks to the efforts of University of South Florida scientists who have isolated and tested an extract from a sponge found in Antarctica.

Tuesday, May 12, 2015

Treatment reduces risk of recurrence of C. difficile infection

In continuation of my update on metronidazole  &  vancomycin

Among patients with Clostridium difficile infection (CDI) who recovered following standard treatment with the antibiotics metronidazole or vancomycin, oral administration of spores of a strain of C difficilethat does not produce toxins colonized the gastrointestinal tract and significantly reduced CDI recurrence, according to a study in the May 5 issue of JAMA.

C difficile is the cause of one of the most common and deadly health care-associated infections, linked to 29,000 U.S. deaths each year. Rates of CDI remain at unprecedented high levels in U.S. hospitals. Clinical infection also has a recurrence rate of 25 percent to 30 percent among affected patients. Not all strains of C difficile produce toxins. Nontoxigenic C difficile strains that lack the genes for toxin production are also found in the hospital environment and can colonize hospitalized patients, although patients are usually asymptomatic. Gastrointestinal colonization by these nontoxigenic C difficilestrains (in both humans and hamsters) has shown promising results as a potential way to prevent CDI, according to background information in the article.

Dale N. Gerding, M.D., of the Edward Hines Jr. VA Hospital, Hines, Il., and Loyola University Chicago, Maywood, Il., and colleagues randomly assigned 173 adult patients who were diagnosed as having CDI (first episode or first recurrence) to receive 1 of 4 treatments: oral liquid formulation of nontoxigenic C difficile strain M3 (VP20621; NTCD-M3), 104 spores/d for 7 days (n = 43), 107 spores/d for 7 days (n = 44), 107 spores/d for 14 days (n = 42), or placebo for 14 days (n = 44). Prior to enrollment, these patients had all successfully completed treatment with metronidazole, oral vancomycin, or both at 44 study centers in the United States, Canada, and Europe.

Thursday, October 2, 2014

Scripps Research Institute Chemists Modify Antibiotic to Vanquish Resistant Bacteria

Scientists at The Scripps Research Institute (TSRI) have devised a new antibiotic based on vancomycin that is powerfully effective against vancomycin-resistant strains of MRSA and other disease-causing bacteria.
The new vancomycin analog appears to have not one but two distinct mechanisms of anti-microbial action, against which bacteria probably cannot evolve resistance quickly.
“This is the prototype of analogues that once introduced will still be in clinical use a generation or maybe even two generations from now,” said Dale L. Boger, the Richard and Alice Cramer Professor of Chemistry at TSRI.
The report by Boger and members of his laboratory was published recently online ahead of print by the Journal of the American Chemical Society.
Increasing Reports of Resistance
Vancomycin entered clinical use in 1958, five years after its isolation from microbes in a soil sample gathered by an American missionary in Borneo. For nearly six decades it has been useful against a wide range of bacteria, and it remains a standard weapon against methicillin-resistant Staphylococcus aureus (MRSA), a major cause of hospital-acquired infections. A compound closely related to vancomycin also has been widely used to protect livestock.
Since the late 1980s, there have been increasing reports of vancomycin resistance in classes of bacteria that usually succumb to the antibiotic, including MRSA. Although vancomycin remains useful, scientists have been looking for new drugs to replace it in cases—often life-threatening—where it no longer can help patients.
The Boger laboratory has focused on inventing improved versions of vancomycin rather than entirely new compounds. “Vancomycin has lasted in clinical use for more than 50 years, in part because it isn’t very vulnerable to antibiotic resistance,” Boger said. “Our thought has been that if we find a vancomycin analog that addresses this current source of resistance we’ll get another 50 years of use out of it.”
Vancomycin works by binding to the building blocks of bacterial cell walls, in a way that prevents their proper assembly and leaves bacteria too leaky to live and replicate. The resistance comes from a single amino-acid alteration that some bacteria make to those building blocks, so that the antibiotic molecule can no longer get a firm grip. That drops vancomycin’s potency by a factor of about 1,000.
‘Incredibly Potent’
In 2012, Boger and his team reported making a vancomycin analog—informally termed vancomycin amidine—with a subtly altered binding pocket that fastens about equally well to the original and resistant sites on bacterial cell wall subunits. To get the precise structural modification they needed, they had to come up with a method for the “total synthesis” of this vancomycin-based compound—a controlled, step-by-step construction using organic chemistry reactions in the lab, rather than a natural enzyme-mediated production within cells.
“Years of work in this lab culminated in a total synthesis strategy that not only allowed us access to this target compound, but also gave us the ability to perform almost any other chemical modification of vancomycin that we wished,” said Akinori Okano, first author of the new report, who is an assistant professor of chemistry at TSRI.
Vancomycin amidine turned out to have acceptable level of activity against vancomycin-resistant and -sensitive bacteria, yet there was room for improvement. Thus in the new study, Okano, Boger and their colleagues used their vancomycin synthesis methods to add an additional feature to the molecule—a peripheral chlorobiphenyl (CBP), long known as a general booster of vancomycin’s potency.
“To our delight, the combination of these modifications led to an incredibly potent molecule, well beyond anything we had expected,” said Okano.


Sunday, June 1, 2014

FDA Approves Dalvance (dalbavancin) to Treat Skin Infections

The U.S. Food and Drug Administration today approved Dalvance (dalbavancin), a new antibacterial drug used to treat adults with skin infections.
Dalvance is intended to treat acute bacterial skin and skin structure infections (ABSSSI) caused by certain susceptible bacteria like Staphylococcus aureus (including methicillin-susceptible and methicillin-resistant strains) and Streptococcus pyogenes. The treatment is administered intravenously.
Dalvance is the first drug designated as a Qualified Infectious Disease Product (QIDP) to receive FDA approval. Under the Generating Antibiotic Incentives Now (GAIN) title of the FDA Safety and Innovation Act, Dalvance was granted QIDP designation because it is an antibacterial or antifungal human drug intended to treat serious or life-threatening infections.
“Today’s approval demonstrates the FDA’s commitment to encouraging increased development and approval of new antibacterial drugs, providing physicians and patients with important new treatment options,” said Edward Cox, M.D., M.P.H, director of the Office of Antimicrobial Products in the FDA’s Center for Drug Evaluation and Research.
As part of its QIDP designation, Dalvance was given priority review, which provides an expedited review of the drug’s application. Dalvance’s QIDP designation also qualifies it for an additional five years of marketing exclusivity to be added to certain exclusivity periods already provided by the Food, Drug and Cosmetic Act.
Dalvance’s safety and efficacy were evaluated in two clinical trials with a total of 1,289 adults with ABSSSI. Participants were randomly assigned to receive Dalvance or vancomycin, another antibacterial drug. Results showed Dalvance was as effective as vancomycin for the treatment of ABSSSI.
The most common side effects identified in the clinical trials were nausea, headache and diarrhea. In the trials, more participants in the Dalvance group had elevations in one of their liver enzyme tests. The Dalvance drug label provides recommendations on dosage adjustment in patients with renal impairment.

Wednesday, June 26, 2013

FDA Approves Vibativ for Hospitalized Patients with Bacterial Pneumonia

We know that, Telavancin (trade name Vibativ) is a bactericidal lipoglycopeptide for use in MRSA or other Gram-positive infections. Telavancin is a semi-synthetic derivative of vancomycin. The FDA approved the drug in September 2009 for complicated skin and skin structure infections (cSSSI)...




Now U.S. Food and Drug Administration today expanded the approved use of the antibiotic Vibativ (telavancin) to treat patients with hospital-acquired and ventilator-associated bacterial pneumonia (HABP/VABP) caused by Staphylococcus aureus. Vibativ should be used for the treatment of HABP/VABP only when alternative treatments are not suitable...

Wednesday, January 2, 2013

Thursday, October 4, 2012

Less commonly prescribed antibiotic may be better for bloodstream infections

 In continuation of my update on Vancomycin

Kevin Chan, MD (Fresenius Medical Care North America and Massachusetts General Hospital) and his colleagues compared the effectiveness of various antibiotics at preventing hospitalization and death from bloodstream infection. They reviewed more than 500,000 blood culture results from their chronic kidney disease database, looking for methicillin-sensitive strains of S. aureus bloodstream infection. They also identified when physicians used vancomycin or cefazolin to treat these infections. Vancomycin is often perceived as the better antibiotic because it has broad coverage against many strains of bacteria; however, other factors like the antibiotic's killing power and tissue penetration are also important factors in selecting the best treatment.


  
Among the major findings:
  • 56% of patients remained on vancomycin after blood culture results reported S. aureus bacteria were susceptible to cefazolin, while only 17% were treated with cefazolin. 
  • Cefazolin-treated patients experienced a 38% lower rate of hospitalization and death compared with vancomycin-treated patients. 
  • Cefazolin-treated patients also had a 48% lower rate of sepsis, which is the most serious form of bloodstream infection.
  • "I think the data suggest there is an opportunity to improve outcomes for patients through appropriate antibiotic selection," said Dr. Chan.

Ref : http://jasn.asnjournals.org/content/early/2012/08/15/ASN.2012010050

Friday, September 9, 2011

Redesigned Vancomycin As Potent Antimicrobial Activity Against Vancomycin-Resistant Bacteria...


In continuation of my update on vancomycin....
A team of scientists from The Scripps Research Institute has successfully reengineered an important antibiotic (Vancomycin)  to kill the   deadliest antibiotic-resistant bacteria. The researchers claim that compound could one day be used clinically to treat patients with life-threatening and highly resistant bacterial infections. The compound synthesized is  an  analogue of the well-known commercial antibiotic vancomycin.

Vancomycin normally works by grabbing hold of and sequestering the bacterial cell-wall making machinery, a peptidoglycan (carbohydrate and peptide containing molecule). Only Gram-positive bacteria have a cell wall, which is a membrane on the cell's outer surface. Unfortunately, bacteria have found a way to alter the peptidoglycan in such a way that the antibiotic can no longer grab hold. Researchers claim that,  the new vancomycin analogue can grab hold of the mutant peptidoglycan, and again prevent the bacteria from making the cell wall and killing the resistant bacteria. But what is so remarkable about the design is that the redesigned antibiotic maintains its ability to bind the wild type peptidoglycan as well.

New compound has an amidine (an iminium, RC=NH+ linked to a nitrogen, N) instead of an amide at a key position buried in the interior of the natural product. I appreciate the idea and the simplicity in achieving the target functional group.

Researchers add that, although it is still at its early stages and there is much work ahead.In my opinion it is a good beginning...



Wednesday, July 21, 2010

How honey kills bacteria..........

We know that, honey has antibiotic activity and has been used specially in burn injuries. Now researchers lead by Dr.Sebastian A.J. Zaat, of Department of Medical Microbiology at the Academic Medical Center in Amsterdam, have come up with an explanation for this antibiotic activity of honey. This first explanation to explain how honey kills bacteria. Specifically, the research shows that bees make a protein that they add to the honey, called defensin-1, which could one day be used to treat burns and skin infections and to develop new drugs that could combat antibiotic-resistant infections.

"We have completely elucidated the molecular basis of the antibacterial activity of a single medical-grade honey, which contributes to the applicability of honey in medicine," said Dr. Sebastian A.J. Zaat...


To make the discovery, Dr. Zaat and colleagues investigated the antibacterial activity of medical-grade honey in test tubes against a panel of antibiotic-resistant, disease-causing bacteria. They developed a method to selectively neutralize the known antibacterial factors in honey and determine their individual antibacterial contributions. Ultimately, researchers isolated the defensin-1 protein, which is part of the honey bee immune system and is added by bees to honey. All bacteria tested, including Bacillus subtilis, methicillin-resistant Staphylococcus aureus, extended-spectrum β-lactamase producing Escherichia coli, ciprofloxacin-resistant Pseudomonas aeruginosa, and vancomycin-resistant Enterococcus faecium, were killed by 10–20% (v/v) honey, whereas 40% (v/v) of a honey-equivalent sugar solution was required for similar activity.


After analysis, the scientists concluded that the vast majority of honey's antibacterial properties come from that protein. This information also sheds light on the inner workings of honey bee immune systems, which may one day help breeders create healthier and heartier honey bees.

http://www.fasebj.org/cgi/content/abstract/24/7/2576

Monday, May 31, 2010

Plectasin - a new weapon against highly resistant microbes ?..

We know that Plectasin, found in Pseudoplectania nigrella (see picture), is the first defensin to  be isolated from a fungus. Plectasin has a chemical structure resembling defensins found in spiders, scorpions, dragonflies and mussels. In laboratory tests, Plectasin was especially active in inhibiting the growth of the common human pathogen Streptococcus pneumoniae, including strains resistant to conventional antibiotics. Plectasin has a low toxicity in mice, and cured them of peritonitis and pneumonia caused by S. pneumoniae as efficiently as vancomycin and penicillin, suggesting that it may have therapeutic potentia.

Now researchers lead by Prof. Dr. Hans-Georg Sahl of   Universities of Bonn, Utrecht, Aalborg and of the Danish company Novozymes AS have shed light on how the substance Plectasin,  destroy highly resistant bacteria. As per the claim by the researchers Plectasin binds to a cell-wall building block called lipid II and thus prevents it from being incorporated and thus disrupting the forming of the cell wall in bacteria so that the pathogens can no longer divide. 

In this process, plectasin behaves like a thief which steals the stones off a mason. 'It binds to a cell-wall building block called lipid II and thus prevents it from being incorporated ,' Professor Sahl explains. 'However, bacteria cannot live without a cell wall.' It comes as no surprise that the most famous antibiotic penicillin also inhibits cell-wall synthesis...
Researchers claims that, plectasin is more similar in its mode of action to another widely used drug, vancomycin. Vancomycin had been the drug of choice in combating MRSA strains since the 1980s. Meanwhile, though, there are more and more bacteria that are also resistant to vancomycin. 'However, these strains are still susceptible to plectasin,' Dr. Tanja Schneider emphasises. Nevertheless, there is no permanent solution to the resistance problem even with a new antibiotic . 'It is always just a question of time until the pathogens mutate and become insensitive ,' she says. 'It's a never ending arms race..' authors conclude that plectasin will be promising lead compound for new antibiotics...

Ref : http://www.sciencemag.org/cgi/content/abstract/328/5982/1168

Tuesday, December 8, 2009

Discovery Of Novel New Class of Antimicrobial Agents... ......

We know that most of the bacteria are getting resistant to the present drugs and there is an urgent need to find a solution for resistant bacteria. Inn this global fight against resistant bacteria many companies are trying different ways and now Chaperone Technologies, Inc has come up with something innovative and interesting way, i.e., the company is trying to develop antimicrobial compounds that work by inhibiting bacterial hsp70 proteins (an entirely new mechanism of action).

Chaperone’s antimicrobial program focuses on development of peptide as well as small molecule hsp70 inhibitor drugs that block the effect of this important class of molecular “chaperones” whose role is to help mediate or respond to toxic misfolded proteins within bacteria. Inhibition of this critical bacterial protein has been proven to kill bacterial pathogens. Besides antimicrobials, the inhibition of hsp70 molecular chaperone proteins present in other cell-types has a range of therapeutic applications that are being investigated by the company.

Using sophisticated computerized molecular modeling techniques, proprietary high-throughput screening tools developed by Chaperone and other approaches, the company has significantly expanded its library of novel hsp70 inhibitor compounds including CHP-267 and CHP-281, just two of the many promising drug candidates from this highly promising family of small molecule inhibitors discovered by the Company. Chaperone is looking at hsp70 inhibitors as stand alone antimicrobial agents as well as in combination with other antimicrobials (e.g., Finafloxacin.HCl : see the structure -which is under phase II clinical trials). The company recently received a US Patent covering a method of significantly amplifying the effectiveness of other antimicrobials by combining their use with that of an hsp70 inhibitor. Combining a bacterial hsp70 inhibitor with another antimicrobial yields increased bacterial killing of clinically important pathogens and the potential for combination therapy.

Chaperone’s drug candidates have been proven effective against dangerous bacteria such as MRSA, acinetobacter, and vancomycin resistant enterococci. When combined with other antibiotics, Chaperone’s compounds stimulate powerful antibiotic synergy, providing superior efficacy even while using significantly lower doses of the combined agents.

Source : http://www.biospace.com/news_story.aspx?NewsEntityId=118501

Tuesday, November 10, 2009

Results of Phase 3 study of fidaxomicin...


We know that Fidaxomicin (also known as lipiarmycin, lipiarmycin A3, tiacumicin B, clostomicin B1, and OPT-80) is a new narrow spectrum macrocyclic antibiotic drug and being developed by Optimer Pharmaceuticals for treatment of Clostridium difficile infection. It works by inhibiting the bacterial enzyme RNA polymerase. It is active against gram positive bacteria especially clostridia.

Recently, Optimer Pharmaceuticals, Inc. (2. Nov, 2009) announced the presentation of new data from fidaxomicin's North American phase 3 study at the 47th Annual Meeting of the Infectious Diseases Society of America (IDSA) in Philadelphia, PA. The results are really encouraging and as per the claim by the company fidaxomicin is associated with faster resolution of diarrhea. In patients with more pronounced diarrhea (ie. not resolving in the first 24 hours of therapy), fidaxomicin was associated with a faster time to resolution of diarrhea than vancomycin (79 hrs vs. 105 hrs). The company claims that the faster time to resolution of diarrhea and improved outcomes for patients requiring concomitant antibiotics are important factors for physicians to consider when selecting a treatment for CDI more interesting factor is the significantly lower recurrence rate and higher global cure rate..

Ref : http://www.optimerpharma.com/news.asp?news_story=100&page_num=