Recognizing High Risk Traumatic Wounds and Preventing Infections and Complications

Review Article / Emergency Medicine and Trauma Care Journal

by Gerry L. Klein and Peter C. Johnson, MedSurgPI, LLC


Abstract: Simple traumatic wounds are a frequent event that can usually be managed without sequelae, unless the wound is of high risk. High risk wounds have a greater propensity to become infected and complicated. Such wounds are characterized by a specific type of wound (i.e. jagged), location of the wound (i.e. lower leg); and patient’s underlying medical condition (i.e: diabetes).If these wounds become infected, they have a negative impact on morbidity, mortality, quality of life, and costs. The take-away should be a wake-up call to physicians specifically and healthcare professionals more broadly that a much more aggressive and effective treatment regimen to prevent wounds from becoming infected is required. Such a regimen should likely include a comprehensive understanding of wound types, the degrees of microbial contamination, and novel ways to prevent infections through wound debridement and irrigation.

Link to read the entire article

Dr. Gerry Klein to serve as panelist on Webinar: Wound Infection in Humanitarian Emergencies


group picture on wound infection Humanitrian Emergencies.jfif

 #PrepareToProtect #IRPWebinar #MedicalResponse #HumanitarianEmergencies #GFARC #WHO  

 We at MedSurgPI, LLC are delighted to participate in and invite you to a webinar on wound infection, organized by Infection Reduction Partners, an initiative to promote and champion cross-sector collaboration and partnerships towards the effective reduction of infection rates worldwide. 

 This webinar will explore the cross-sector data, guidelines, and best practices in wound infection during disaster response and humanitarian emergencies. A presentation of practical and innovative options to reduce the risk of infection will be shared and we will discuss current publications including IFRC’s “International First Aid, Resuscitation and Education Guidelines 2020,” WHO’s Guidance, “Prevention and Management of Wound Infection,” WHO/ICRC’s “Management of Limb Injuries,” and WHO’s “Global Guidelines for the Prevention of Surgical Site Infection”.

 Panelists: 

Dr. Pascal Cassan, MD, Head of Global First Aid Reference Centre, International Federation of Red Cross and Red Crescent Societies
Flavio Salio, Team Lead - Emergency Medical Teams at the World Health Organization
Kim Delahanty, BSN, PHN, MBA/HCM, CIC, FAPIC, Infection Prevention Control Advisor/Referent, Médecins Sans Frontieres USA/OCP (Doctors without borders)
- Dr. Gerald L. Klein, MD, Principal at MedSurgPI, LLC, wound infection consultant 

Moderator: 

Tessy Antony de Nassau, Ambassador for UNAIDS, and Patron to UNA-UK

 Find out more on IRP social media FacebookYouTubeTwitterInstagramLinkedIn!   

Audience: 

  • Medical team members involved in humanitarian aid, disaster relief, and emergency response: Medical directors, doctors, nurses, technicians, responders, infection preventionists, health officers, medical team leaders

  • NGOs

  • International Organizations

  • Government/Ministries of Health

The Medical Monitor is one of the key players in keeping subjects safe in clinical trials. This article helps explore the role of the Medical Monitor

Journal of Clinical Research Best Practices

Medical Monitoring of Clinical Research Studies

By Gerald L. Klein, Peter C. Johnson, and Roger Morgan

Introduction

The Medical monitor’s (MM’s) primary responsibilities in a clinical trial are to oversee the safety and protection of the research subjects and to provide independent oversight to help ensure the scientific reliability, clinical integrity, and quality of the clinical trial. Although the Food and Drug Administration (FDA) has not spelled out the necessity and role of the medical monitor, MM participation is important for compliance with Good Clinical Practice (GCP) guidelines and MMs almost always play a significant role in multicenter clinical trials.1 The best practices for MMs described in this article can also help study sponsors comply with International Council for Harmonization (ICH) guidelines, Technical Requirements for Pharmaceuticals for Human Use, the United States Code of Federal Regulations (CFR), and FDA regulations and guidances.2

The MM collaborates with the project manager, safety, data management, biostatistics and quality departments, principal and sub-investigators, the site coordinator, the Data & Safety Monitoring Board (DSMB), and any other group that directly or indirectly protects the safety of study participants.

Communications

Prompt and transparent communication is an essential element underlying safety in all human clinical studies.3 The MM should be the point person for medical, scientific and safety questions posed by clinical investigators, their site personnel, and staff at the study sponsor and contract research organization (CRO) involved in the trial. The MM’s contact information should be readily available to investigators and their staff. The MM should be available essentially 24/7 with a back-up MM when the primary MM is not available.

MMs often address significant questions on the following topics:

  • Documentation

  • Protocol and investigator brochure

  • Inclusion criteria

  • Exclusion criteria

  • Safety

  • Patient concerns

  • Adverse events (AEs)

  • Serious adverse events (SAEs)

  • Suspected Unexpected Serious Adverse Reactions (SUSAR)

  • Pregnancy

  • Medication errors

  • Concomitant medications

  • Laboratory values

  • Protocol deviations and waivers

  • Informed consent

  • Unblinding

  • Early termination or withdraw of a subject

  • Protocol-stopping rules

  • Other topics related to clinical research

  • DSMB and pharmacovigilance team questions with respect to adverse events

MMs document all relevant communications in the appropriate database. When there are multiple significant errors at an investigational site, the MM may be called into an investigation to determine the cause and whether corrective actions or new training is required.4 In rare cases, the MM, together with the project manager and quality assurance, assesses whether a site must be removed from a study due to safety issues, poor data quality, or violation of GCPs.

Maintain a Question and Answer (Q & A) log. Create anticipated Q & A’s prior to the study and then maintain a log to help provide quick, accurate and consistent answers to repeat questions. The clinical sites should be able to search the log for themselves.

Protocol and Investigator’s Brochure (IB)

The MM may write all or just parts of the protocol and investigator’s brochure. At minimum, the MM should review and approve these documents.5 Since the MM is expert on the protocol and the specific therapeutic indication being studied, the best practice is to involve the MM in training sponsor and/or CRO staff as well as the investigators and their personnel on the protocol and IB.

The MM should ensure that protocol endpoints make medical and scientific sense and are safely achievable. The clinical trial’s expected benefits must outweigh its risks.6 Inclusion and exclusion (I/E) criteria must align with this goal. I/E criteria must prevent the enrollment of subjects who are unlikely to obtain a positive therapeutic clinical endpoint or would be put at unacceptable risk in the study. For instance, a patient with a childhood history of bronchial asthma may not be appropriate to enroll in a clinical trial testing a medication that has properties of beta blockers, which can exacerbate symptoms of asthma.

The MM reviews permitted and prohibited concomitant medications (including over-the- counter drugs, herbs and dietary supplements) for possible interactions with the molecule being studied. The MM also ensures that the protocol does not specify types or numbers of procedures that would pose unnecessary risks for study subjects. The MM may recommend ways mitigate such risks.

The MM ensures that the IB clearly describes non-clinical studies and any adverse events of special interest (AESIs). An example of an AESI would be an abnormal electrocardiogram when all cardiac adverse events are of special interest to the regulatory authorities. All current knowledge about the drug, device or biologic must be clearly spelled out in the IB, not hidden in esoteric study reports.7 It is unfortunate that many investigators do not read the IB in detail. Therefore, the MM should try to convey the important aspects of the pharmacokinetics, pharmacodynamics, metabolism, drug interactions and expected adverse events associated with the study therapy to the investigator and the appropriate staff at the clinical site.

Review and Discussion

The MM reviews each subject’s eligibility data, screening physical examination results, medical history, concomitant medications, and laboratory tests before approving their entry into the study. If the investigator is attempting to enroll unqualified subjects, training may be required. The MM thoroughly discusses non-trivial protocol deviations (PDs), which should be rare, with the investigator, and only the most minor ones should be approved. A major PD may affect subject safety, data integrity, or the integrity of the entire study.8 A dosing error by which a subject received twice the dose of the investigational drug during one dosing interval would be a major deviation. A subject’s labs being a few hours out of the visit window would be a trivial deviation.

If time permits, the investigator should consult with the MM before unblinding a subject so the MM can assess and document the decision. The investigator should also discuss with the MM any early unusual termination or withdrawal of a subject from the study.

Data & Safety Monitoring Board (DSMB) or Data Safety Committee (DSC)

If there is a DSMB or DSC, the MM should participate in the blinded section of any meetings to help answer any questions related to adverse events and other potential safety and enrollment issues.9

Adverse Events

One of the most significant MM responsibilities is to work with the investigator to determine the most accurate causality of Serious Adverse Events (SAEs) and Suspected Unexpected Serious Adverse Reactions (SUSARs). SUSAR expectedness determinations should be based on the Reference Safety Information section of the IB, or, in studies of marketed drugs, the applicable package insert. Many investigators do not have a good understanding of causality assessment, and poorly defined regulatory terms and examples do not help the situation.10 Following the CIOMS report recommending a binary approach of “related” or “not related” in determining causality simplifies the complex and confusing terminology.11 There is no accepted standard for assigning causality to an SAE, but employing the following Bradford Hill Criteria is an excellent way to determine causality:12,13

1.       Strength of Association. A strong association between a treatment and an adverse event indicates causation. For example, each time the drug was given to a subject, it caused vomiting within a predictable time period.

2.       Consistency. Established adverse event attributions or previous determinations in similar situations indicate causation.

3.       Specificity. An established mechanism of action connecting the treatment and the adverse event indicates causation.

4.       Temporality. Exposure to the product must occur before the disease or event, and not after a latency period. However, temporality is not sufficient to establish causation.

5.       Biological Gradient. A dose response effect is a strong argument for causation.

6.       Plausibility. The causal relationship is biologically plausible.

7.       Coherence. The known facts fit the natural history and biology of the disease.

8.       Experiment. Epidemiologic studies indicate causation.

9.       Analogy. A similar agent causes the same type of AE.

 Safety and Pharmacovigilance Reporting

The MM develops or reviews a brief narrative describing each SAE and SUSAR, which should include the following elements:14

  • Clinical event (postmortem findings if applicable)

  • Course of event, with temporal relationship to experimental product

  • Outcome of the event with the nature, severity and intensity

  • Relationship of the subject’s medical history and concomitant medications to the event

  • Significant test results or laboratory findings

  • Therapeutic treatment for the event

  • Action, if any, taken with regard to experimental product

  • Causality assessment by investigator and sponsor

  • Review and analysis of similar events with the experimental product

The MM works with the safety/pharmacovigilance team to code adverse events based on the latest edition of the Medical Dictionary for Pharmaceuticals for Human Use (MedDRA).15 The MM also reviews the coding of concomitant medications using the World Health Organization’s (WHO’s) latest edition of pharmaceutical names (when it is used in the clinical trial). The MM reviews all SAE and SUSAR reports for accuracy and completeness and is the point person to discuss such events with the sites.

 Conclusion

Since there are no regulatory guidelines on MM duties, this article has discussed those that are most significant. With the possible exception of the lead principal investigator, the MM should be the person most expert on the medical and scientific aspects of a multicenter study. The principal investigator at each site and the MM, along with DSMB and the institutional review board (IRB), share primary responsibility for the health and safety of study subjects and ensure the validity of the study. They must establish working relationships to ensure that subjects are protected and study data, including SAE and SUSAR reports, are accurate. This profound responsibility means that the MM for a study must have the requisite expertise, personality, dedication and ability to use the processes outlined in this article.

 References

  1. Vijayananthan A. et al., “The importance of Good Clinical Practice guidelines and its role in clinical trials.” Biomed Imaging and Intervention Journal, 2008; 1: e5.

  2. E6(R2) Good Clinical Practice: Integrated Addendum to ICH E6 (R1) Guidance for Industry, FDA, March 2018.

  3. “FDA Guidance for clinical investigators, sponsors, and IRBS: adverse event reports to IRBs-Improving human subject protection,” FDA, Jan 2009.

  4. Knepper D. et al., ‘Detecting data quality issues in clinical trials: current practices and recommendations,’ Therapeutic Innovation 7 Regulatory Science, 2016; 50:15- 21.

  5. World Health Organization, Guide for writing a Research Protocol for research involving human participation; 2014.

  6. Emanuel E. et al., “What makes clinical research ethical?” JAMA, 2000; 283:2701- 2711.

  7. Fiebig D. et al., “The investigator’s brochure: A multidisciplinary document,” Medical Writing, 2014; 23:96-100

  8. Attachment C: Recommendation on Protocol Deviations, Office of Human Research Protection, HHS.gov.

  9. Calis A. et al., “Understanding the functions and operations of data monitoring committees: survey and focus group findings,” Clinical Trials, 2017:59-66.

  10. Morse M. et al., “Monitoring and ensuring safety during clinical trials,” JAMA. 2001; 285:1201-1205

  11. “Management of Safety Information for clinical trials: Report of CIOMS VI,” 2005.

  12. Federak, K. et al., “Applying the Bradford Hill criteria in 21st century data integration has changed causal inference in molecular epidemiology.,” Emerging Themes in Epidemiology, 2015; 12:14-23

  13. Howick, J., “The evolution of evidence hierarchy: What can Bradford Hill’s guidelines of causation contribute?” Journal for the Royal Society of Medicine, 2009; 102; 2009:186-194

  14. Modified from Ledade, S. et al., “Narrative writing: Effective ways and best practices. Perspectives in Clinical Research,” Perspectives in Clinical Research, 2017; 8:58-62

  15. Brown. E. G., “Using MedDRA: implications for risk management,” Drug Safety. 2004;27(8):591-602

Authors

Gerald L. Klein, MD, is a principal at MedSurgPI. Contact him at 1.919.930.9180 or gklein@medsurgpi.com.

Peter C. Johnson, MD, is president and CEO of Cell X Technologies.

Roger Morgan, MD, is vice president of medical affairs at MedSurgPI, LLC.

Improving Clinical Trial Safety

There is a dire need to establish transparent medication safety guidelines and to assure the public of the industry’s commitment to this task.  Clinical investigators and their staffs who conduct clinical trials often do not understand some of the regulatory methodology used to assess medication safety, particularly with regard to the assignment of causality to drug-related Serious Adverse Events.  Yet, this is critical to establish the safety of medication.   The medical monitor in a clinical trial can help educate and guide investigators in a clinical study with regard to accepted causation assignment.  Since there is a paucity in the literature regarding medical monitoring of clinical trials, this article will help to fill this gap, and to provide a guide to establishing proper causality of an adverse event.  This open access article can be found at “Medical Monitoring of Clinical Research Studies.

MAINTAINING PRODUCT DEVELOPMENT MOMENTUM IN A CHALLENGING TIME

It is important that biotech and device companies continue their critical research and development during these chaotic times.  Many small companies are having difficulty raising funds and may be getting short of cash.  Consulting companies have the flexibility to  work remotely with these companies and can  consider delayed payments, use of equity, or other innovative means to compensate them for their assistance while keeping momentum strong and morale high.  MedSurgPI, LLC fulfills this role by providing virtual medical support for product development, medical monitoring, medical affairs, and serving as Chief Medical Officers.  Should you require these services, please contact us at the following:

Drugs and Biologicals:  gklein@medsurgpi.com    

Devices:  pjohnson@medsurgpi.com

Medical Monitoring and DSMB activity:  rmorgan@medsurgpi.com

Find us at: www.medsurgpi.com

MedSurgPI, LLC provides remote Medical Officer and Commercial Chief Medical Officer Services during this critical time of the Covid-19 Outbreak.

Social distancing during the Covid-19 crisis is adversely affecting the ability of companies to continue biomedical product development and to plan and execute clinical trials and medical affairs activities. While this crisis will eventually abate (albeit with an uncertain timeline) virtually located professionals are needed in the interim to continue to support these critical development activities.  Research Triangle Park, NC-based MedsurgPI, LLC offers 20+ experienced Physician Associates from all areas of Medicine to provide comprehensive remote medical officer and medical monitoring  services to companies worldwide.  These services also include strategic product development oversight, clinical trial planning, Contract Research Organization sourcing and management,  Medical Communications, interim management, support for fundraising, Key Opinion Leader recruitment and facilitation and medically relevant corporate representation.  Should your organization require these skills on a remote basis in the near future, please contact MedsurgPI, LLC and we will respond immediately to support you.

Peter C. Johnson, MD and Gerald L. Klein, MD

Co-Principals, Medsurgpi, LLC

www.medsurgpi.com

pjohnson@medsurgpi.com

gklein@medsurgpi.com

 

Australian R&D Tax Incentive

How The Australian Research and Development (R&D) Tax Incentive Works

Peter C. Johnson, MD and Gerald L. Klein, MD

Principals, MedSurgPI, LLC

Introduction

In order to foster greater corporate R&D investment to spur jobs and innovation, the Australian government adopted the R&D Tax Incentive system in 2011.  While its stipulations have gradually changed over the years, it remains a generous incentive for companies large and small to invest in R&D in Australia.  The plan has two broad elements, one aimed at supporting companies having less than 20 million in annual revenue (these are Australian dollars, equivalent to 13.4 million US dollars) and those having greater than 20 million in revenue.  The incentive applies not only to native Australian companies but also to foreign companies that establish a bona fide Australian presence and comply with Australian business guidelines as set by the Australian Securities and Investments Commission (ASIC: https://www.investopedia.com/terms/a/australian-securities-and-investments-commission-asic.asp).  A straightforward set of FAQs regarding these requirements can be seen here: http://cosec.com.au/frequently-asked-questions/faqs-on-australian-companies/.   Additionally, types of R&D activities that are eligible for the benefit are carefully defined in order to prevent abuse of the system.  These and current applications of the Incentive are outlined below:

R&D Expenditure Threshold

For all companies, the threshold R&D spending amount to which the Incentive applies is now 150M Australian dollars (100.5 million US dollars).

Companies Having Less Than 20 Million in Revenue

Such companies are entitled to a refundable tax credit equal to their corporate tax rate percentage plus 13.5 basis points.  Given that the present corporate tax rate is 27.5%, their refundable tax credit as of the 2019 legislation is 41% of their R&D spend.  The word “refundable” requires some explanation.  For companies whose revenues impart no tax burden, they are entitled to a cash rebate of 41% of their R&D spend.  If they have a tax burden, the 41% will initially be used to offset that amount of tax burden up to and including 41% of their R&D spend.  If their tax burden does not achieve this level, the refundable incentive will first be used to offset their tax burden and the remainder distributed to the company as a cash rebate.

The Incentive also distinguishes between expenditures for clinical trials and for R&D that is not specifically a clinical trial expenditure (such as preclinical studies).  There is a 4 million Australian dollar (2.68 million US dollar) cap on the latter activities but no limit on the Incentive as applied to clinical trial activities up to the R&D Expenditure Threshold of 150 million Australian dollars.

Some examples are shown in as follows, applicable only to companies having less than 20 million Australian dollars in revenue.

Example 1: Acme Medical spent zero dollars on Clinical R&D and six million dollars on Non-Clinical R&D.  Its tax burden was zero so its tax offset is zero and it receives a cash rebate of four million dollars.*

Example 2: RNA Science spent one hundred million dollars on Clinical R&D and zero dollars on Non-Clinical R&D.  Its tax burden was zero so its tax offset is zero and it receives a cash rebate of forty one million dollars.

Example 3: Mitochondrial Solutions spent zero dollars on Clinical R&D and ten million dollars on Non-Clinical R&D.  Its tax burden was four million so its tax offset is four million and it receives a cash rebate of zero dollars.*

Example 4: Atlas Diagnostics spent fifty million dollars on Clinical R&D and six million dollars on Non-Clinical R&D.  Its tax burden was two million so its tax offset is two million and it receives a cash rebate of 19.7 million dollars.**

*Only the first 4M of any R&D tax offset for non-clinical trial related activities can be applied each year.  Any remainder must be carried forward as a non-refundable tax offset (see below) that can be applied in future years.  This limit does not apply to clinical trial activities.

*In this instance, non-clinical R&D spend refund, capped at 4M is partially applied (2M) to offset the 2M tax burden, leaving 50-2=48*.41=19.7M available as a cash rebate.

Companies Having Greater Than 20 Million in Revenue

Circumstances of application of the Incentive are quite different for larger companies.  Companies having greater than 20 million in revenue are entitled to a non-refundable tax incentive equal to their corporate tax rate PLUS a sliding scale premium based on the level of intensity of their R&D expenditure.  “Non-refundable” in this instance means that the Incentive rebate only applies to offsetting the company’s tax burden with no cash rebate.  However, if the calculated tax incentive amount exceeds that of its annual tax burden, the residual amount can be carried forward as an applicable tax credit for future years.

The sliding scale R&D “intensity premium” (that is, above and beyond their corporate tax rate)  is calculated as a function of the percent of a company’s total expenses that are represented by R&D expenses.  For an R&D Intensity Range (that is, R&D expense as a percentage of total expenses) that is less than or equal to 4, the Incentive Premium (% of Total Expense) is 4.5.  When the R&D Intensity Range is greater than 4 but less than or equal to nine, the Incentive Premium rises to 8.5%.  Finally, when the R&D Intensity Range is greater than nine, the Incentive Premium rises to 12.5%.

The following provides some examples of how the program applies to entities having greater than 20 million in annual revenue.  Note that corporations having greater than 50 million in annual revenue are subjected to a 30% corporate tax rate.

Example 1: Claro Science’s R&D expense is 1 million versus total expense of 40 million.  It has 45 million in revenue.  Its applicable corporate tax rate is 27.5%.  Its tax burden is 12.4 million and its R&D expense as a percent of total expense is 2.5.  Its Incentive Premium (% of total expenses) is 4.5.  Its earned incentive tax offset is 18 million.  Therefore, this year its incentive tax offset is 12.4 million and it is able to carry forward 5.6 million in incentive tax offset for future years.

Example 2: Adams Medical’s R&D expense is 3 million versus total expense of 50 million.  It has 60 million in revenue.  Its applicable corporate tax rate is 30%.  Its tax burden is 18 million and its R&D expense as a percent of total expense is 6.  Its Incentive Premium (% of total expenses) is calculated as 4.5(4)+8.5(2).  Its earned incentive tax offset is 35 million.  Therefore, this year its incentive tax offset is 18 million and it is able to carry forward 17 million in incentive tax offset for future years.

Example 3: Hong Kong System’s R&D expense is 9 million versus total expense of 135 million.  It has 150 million in revenue.  Its applicable corporate tax rate is 30%.  Its tax burden is 45 million and its R&D expense as a percent of total expense is 6.7.  Its Incentive Premium (% of total expenses) is calculated as 4.5(4)+8.5(2.7).  Its earned incentive tax offset is 40.3 million.  Therefore, this year its incentive tax offset is 40.3 million and it is able to carry forward zero dollars in incentive tax offset for future years.

Example 4: Pacific Bio’s R&D expense is 50 million versus total expense of 450 million.  It has 500 million in revenue.  Its applicable corporate tax rate is 30%.  Its tax burden is 150 million and its R&D expense as a percent of total expense is 11.1.  Its Incentive Premium (% of total expenses) is calculated as 4.5(4)+8.5(5)+12.5(2.1).  Its earned incentive tax offset is 86.8 million.  Therefore, this year its incentive tax offset is 86.8 million and it is able to carry forward zero dollars in incentive tax offset for future years.

*R&D Expenditure Threshold capped at 150M.  See above.  Also, the scale applies incrementally – that is R&D expenditure up to 4% is rewarded by a 4.5% inventive premium, that portion of R&D expenditure between 4% and up to 9% is rewarded by an 8.5% incentive premium and that portion of R&D expenditures > 9% of total expenses is rewarded by a 12.5% incentive premium.

In Country Guidance

Support for companies seeking these benefits – especially foreign entities with an Australian presence – can be assisted through the complexities of compliance and application by local consultancies having expertise in these areas.  A Google search entitled “Australian firms providing R&D tax incentive support” provides a starting point for the identification of such groups: https://search.yahoo.com/search?fr=mcafee&type=E211US667G0&p=Australian+firms+providing+R%26D+tax+incentive+support.  Australian Contract Research Organizations are also adept at guiding clients to such support organizations.

Overall, the Incentive is jointly administered by Innovation and Science Australia (ISA) and the Australian Taxation Office (ATO), the former responsible for registering R&D activities while the latter manages the rules regarding who is eligible and overseeing acceptable costs.  Background information and guidance regarding eligibility and application to the program can be found here: https://www.business.gov.au/grants-and-programs/research-and-development-tax-incentive

The revised legislation governing the program as introduced into the Australian Parliament in 2019 can be found here: Treasury Laws Amendment (Research and Development Tax Incentive) Bill 2019.

MedSurgPI Partnership Announcement

 
Divicia Image.png

MEDSURGPI PARTNERSHIP ANNOUNCEMENT WITH DEVICIA FOR EU DEVICE EXPERTISE

MedSurgPI is expanding our services through our unique partnerships with experienced, dedicated, entrepreneurial consulting companies. In light of the current significant regulatory changes in the EU, increasing our regulatory and clinical expertise in Europe is essential. For that reason, we are proud to announce our new relationship with Sweden-based Devicia, a full-service medical device exclusive CRO. As members of Technical Committees and nominated as experts in international working groups writing medical device standards (such as ISO 14155), we will be able to provide our clients with an industry perspective and first-hand expert advice.

 Through this collaboration, MedSurgPI will provide:

·       MDR & IVDR transit and compliance  

Devicia has extensive knowledge of regulatory changes currently taking place in the EU with the implementation of medical device regulation (MDR) and in-vitro diagnostic device regulation (IVDR). Through the collaboration, MedSurgPI and Devicia can assist with regulatory strategy & gap analysis, technical files and design dossiers, classifications, regulatory submissions etc.

·       Clinical Evaluations

A key component in getting or maintaining a CE mark is the Clinical Evaluation Report. All medical devices on the EU market must perform a Clinical Evaluation according to MEDDEV 2.7/1 Rev 4. Devicia’s experienced medical writing team performs clinical evaluations according to the current guidelines and fulfils the requirements of the role of a clinical evaluator.

·       EU Device Clinical Investigation

Devicia provides full-service support for all kinds of clinical investigations (single center, multicenter multinational RCT’s, registries etc.) both before and after CE mark. Devicia also supports clients with the new MDR requirements for Post Market Clinical Follow up (PMCF).  Devicia is nominated as a Swedish expert in the ISO/Technical Committee 194 Working Group 4 governing clinical investigations of medical devices in humans.

 ·       Licenses for ISO 14155 Clinical QMS

 ISO 14155:2011 is specifically tailored to the requirements for medical device clinical investigations. Through the collaboration, MedSurgPI can offer access through licenses to SOP’s, templates and forms to be able to conduct a clinical investigation according to ISO 14155:2011.

 gklein@medsurgpi.com                         pjohnson@medsurgpi.com

919-930-9180                                                   919-807-9132

 

The Coming of Age of the Antimicrobial Post-Op Dressing?

Authors:  Gerald L. Klein, MD & Peter C. Johnson, MD

Post-operative home care has become more important than ever.  The use of an antimicrobial dressing in this setting is often essential to an uncomplicated recovery.  Previously, tape and gauze may have sufficed for most incisions and antimicrobial dressings were reserved for those incisions at the highest risk of infection.  However, the earlier discharge of surgical patients puts them at increased risk of acquiring a post-op surgical infection, which is most likely to occur 24-48 hours after surgery.  Increasingly, proper home management of wounds is critical to prevent infections.

As patients become more responsible for their wound care and adherence to follow up instructions becomes more critical, the following questions pertain: “What best post-operative practices reduce the risk of an infection?”  Furthermore, what options exist that empower the patient to be a good partner in their post-op care and recovery?

Dressing Features That Enable the Patient: Non-Invasive, Observational Wound Management

Educated patients with the resources to follow their discharge instructions are more likely to experience a positive outcome than those without.  Once home, the onus is on them to manage the care of their own surgery site.  So, what should the dressing provide to help them be a good partner in their care?

·       The dressing should contain an appropriate concentration of a broad-spectrum antimicrobial to prevent microbial growth at the patient-dressing interface.

·       The dressing should be thin and be attached with an adhesive that can withstand at least a week of wear time without being irritating. 

  • This obviates the need for dressing changes, saves time, cost, decreases skin irritation and reduces the potential for the new introduction of microbes.

·       The filmic portion of the dressing should allow for gas transfer without allowing microbial or fluid entry.

·       The dressing adhesive and film should be sufficiently pliant to conform to irregular wound shapes to form a seal with the skin.

·       The dressing should be transparent to enable both patient and physician to observe the incision for signs of inflammation and/or drainage.

If these criteria are met, the patient is provided with convenient and comfortable post-operative wound care.  Moreover, the ability to readily observe the status of the wound makes the patient a better partner to the physician in the early post-operative period when the potential for Surgical Site Infection is at its highest.

Commonly used antimicrobial dressings typically include one of the following:

·       PHMB (Polyhexanide)

·       Silver

·       CHG (Chlorhexidine gluconate)

·       Silver + CHG

 Lack of Guidelines to Prevent Surgical Site Infections (SSIs)

It is unfortunate that the current guidelines do not yet recommend antibiotic dressings but state that the issue is unresolved.  The earlier surgical hospital discharge should help necessitate this change. 

 Current Guidelines

Guideline & Conclusion

1.     CDC Guideline for Prevention of SSI, 2017 No recommendation/unresolved issue

2.     WHO Global Guidelines on the Prevention of SSI, 2016 Conditional recommendation

3.     American College of Surgeons and Surgical Infection Society Guidelines for Prevention and Treatment of SSI, 2016 No recommendations regarding choice of post-operative dressings.

4.     Society for Healthcare Epidemiology of America/Infectious Disease Society of America: Strategies to Prevent Surgical Site Infection in Acute Care Hospitals: 2014 Update Post-operative wound dressings not addressed

 References:

1.     CDC Guideline for Prevention of SSI, 2017. https://jamanetwork.com/journals/jamasurgery/fullarticle/2623725 

2.     WHO Global Guidelines on the Prevention of SSI, 2016. http://apps.who.int/iris/bitstream/handle/10665/250680/9789241549882-eng.pdf;jsessionid=9AAB8F406232C1294A363043D58CAF6F?sequence=1

3.     American College of Surgeons and Surgical Infection Society Guidelines for Prevention and Treatment of SSI, 2016 https://www.journalacs.org/article/S1072-7515(16)31563-0/fulltext

4.     Society for Healthcare Epidemiology of America/Infectious Disease Society of America: Strategies to Prevent Surgical Site Infections in Acute Care Hospitals: 2014 Update

https://www.jstor.org/stable/10.1086/676022

 One such post-operative wound dressing that fulfills these requirements is Eloquest’s ReliaTect™. 

A phase 1 clinical trial of the sigma-2 receptor complex allosteric antagonist CT1812, a novel therapeutic candidate for Alzheimer’s disease

Alzheimer’s & Dementia: Translational Research & Clinical Interventions 5 (2019) 20-26


Michael Grundmana, Roger Morgan, Jason D. Lickliter, Lon S. Schneider, Steven DeKosky, Nicholas J. Izzo, Robert Guttendorf, Michelle Higgin, Julie Pribyl, Kelsie Mozzoni, Hank Safferstein, Susan M. Catalanog


We would like to congratulate Roger Morgan, MD; for being a co-author on this new publication

Roger is the VP of Medical Affairs at MedSurgPI LLC.  MedSurgPI is a medical consulting group specializing in strategy for product development, medical affairs, medical monitoring, and fractional medical officer services.  For additional information about MedSurgPI services contact gklein@medsurgpi.com

Read the full article

Abstract:

Background: Elayta (CT1812) is a novel allosteric antagonist of the sigma-2 receptor complex that prevents and displaces binding of Ab oligomers to neurons. By stopping a key initiating event in Alzheimer’s disease, this first-in–class drug candidate mitigates downstream synaptotoxicity and restores cognitive function in aged transgenic mouse models of Alzheimer’s disease.

Methods: A phase 1, two-part single and multiple ascending dose study was conducted in 7 and 4 cohorts of healthy human subjects, respectively. In part A, healthy, young subjects (,65 years old) received CT1812 doses ranging from 10 to 1120 mg (6:2 active to placebo [A:P] per cohort). In part B, subjects were administered 280, 560, and 840 mg once daily for 14 days (8:2 A:P per cohort). An elderly cohort, aged 65-75 years, was dosed at 560 mg once daily for 14 days (7:2 A:P). Serum concentrations of CT1812 in part B were measured on day 3 and 14 and cerebrospinal fluid concentrations on day 7 or 9. Cognitive testing was performed in the healthy elderly cohort at baseline and at day 14 of treatment.

Results: Treatment with CT1812 was well tolerated in all cohorts. Adverse events were mild to moderate in severity and included headache and GI tract symptoms. Plasma concentrations of drug were dose proportional across two orders of magnitude with minimal accumulation over 14 days. Cognitive scores in the healthy elderly cohort were similar before and after treatment.

Conclusions: CT1812 was well tolerated with single dose administration up to 1120 mg and with multiple dose administration up to 840 mg and 560 mg in healthy young and healthy elderly subjects, respectively. CT1812 is currently being studied in early phase 2 trials in patients with Alzheimer’s disease.  2018 The Authors. Published by Elsevier Inc. on behalf of the Alzheimer’s Association. This is an open access article under the CC BY-NC-ND license: http://creativecommons.org/licenses/by-nc-nd/ 4.0/



Neglecting Regulations and Quality System Requirements is Detrimental to your Business

Mark E. Ramser, VP Quality Management, MedSurgPI, LLC; Peter C. Johnson, MD, Principal, MedSurgPI, LLC; Gerald L. Klein, MD, Principal, MedSurgPI, LLC

It’s never too early to start designing and implementing a company’s quality management system (QMS). Initiating the R&D process is not a problem for companies that have a well-defined QMS and a mature understanding of how it should operate. However, without a well-defined, compliant and effective system, this could be the start of potential future quality and regulatory nightmares. Even with a well-defined system in place, significant problems can occur if the organization is lacking the maturity and experience to utilize it properly. Typically, the R&D function is where most people think a product starts. The founders may not have any understanding of regulatory or quality system requirements. They can simply be focused on developing and launching a product, choosing to worry about the regulatory and quality system requirements once they have a product to sell. Unfortunately, by waiting until the product is ready for market, it will be too late. The ISO 13485 standard, 21 CFR Part 820 (FDA Regulations) and MDR (European Medical Device Regulations) require and expect the product development and launch to be performed under controlled processes and systems. Refer to the following links for the above referenced standards and regulations:

  • ISO 13485 (Quality Management System for Medical Devices)

  • 21 CFR Part 820 (FDA Medical Device Regulations)

  • MDR (European Medical Device Regulation)

The notified bodies and regulators expect the following areas to be controlled and managed per their documented standards and regulations:

Mark Ramsey graphic.png

The most critical in early stage product development are:

  • Documented Quality Management System

  • Design Controls (Design History File sub-bullets to prove all these areas are addressed)

    • Design and Development Planning

    • Design Inputs

    • Design Outputs

    • Design Reviews

    • Design Verification and Validation

  • Change Management (design, system, process, supplier, raw material, etc.)

  • Process and Production Control

    • Process

    • Equipment

    • Supplier Selection and Control

Many of these activities need to be initiated from the start of the R&D process and the establishment of a Quality Policy and Quality Process at the proper time falls squarely upon the CEO. Companies cannot generally recreate documentation history at later dates in preparation for an FDA or regulatory body audit. The company is then open to all the risks related to not having required systems during product development. The risks can vary based on the severity of the issue and can range from:

  • A Form 483 may be issued to the firm. Form 483 is the communications method used by the FDA to inform the company’s management of objectionable conditions. This should be followed up with a thorough investigation, root cause analysis and corrective action within 15 days. These are available to the public, but only when specifically requested.

  • Severe issues on Form 483 or a significant number of issues on Form 483 could result in the issuance of a Warning Letter. A Warning Letter is one of the FDA’s principal means of achieving prompt voluntary compliance with the Act. The warning letters are much more significant than a Form 483 and are publicly posted on the FDA website and are easily searchable. This must be followed up with a thorough investigation, root cause analysis and corrective action within 15 days.

  • Additional actions can result in a consent decree, product seizure and up to and including criminal prosecution against the firm and individuals, with a special focus on the CEO.  A consent decree may be viewed as the equivalent to a court order under which the manufacturing and distribution of products can only resume, with conditions closely monitored by FDA.

The regulations defined in 21 CFR Part 820 are legal requirements and carry the stiff penalties noted above. This carries much more significance than merely an ISO standard that defines the requirements without having any legal recourse. One must keep this in mind from the earliest possible point of the inception of the company and not just during the product launch. Not following QMS at product inception and all along the development cycle can have dire consequences for the company.

The timing of onset and the complexity of any company’s QMS is dependent upon the risk that a product may represent when used with humans. Therefore, judgment and expertise are required to properly craft and institute such systems. MedSurgPI can assist your company at the earliest stages to institute a QMS, perform Gap Assessment and Corrective Actions for an existing QMS and assist with the ongoing management of a QMS. Please contact MRamser@medsurgpi.com for additional information.