NG Focus Group-Newsletter II · David Putnam, Prabhas Moghe, Chun Wang, Zheng-Rong Lu, Thomas...

13
June 2013, Issue II 2013 AAPS Nanotechnology Focus Group News-letter Message From Chair University Distinguished Professor and Director, Center of Pharmaceutical Biotechnology and Nanomedicine, School of Pharmacy, Northeastern University. Dear Readers, You are keeping in hands the second issue of our New Letter. The enthusiastic team led by Shardool Jain, PhD candidate from Northeastern, has put together another set of materials relating to the activity of the Nanotechnology Focus Group of AAPS. A lot happened over the months since the first issue of News Letter was brought to you. In addition to Physical Pharmacy and Biopharmaceutics (PPB) and Formulation Design and Development (FDD) Sections, Nanotechnology FG became affiliated with Drug Discovery and Development Interface (DDDI) Section of AAPS, which still further strengthen our networking within the AAPS and improved our visibility. Group representatives took active participation in preparation of 2013 and 2014 Biotechnology Conferences and AAPS Annual Meetings, where our FG will be represented by various forums, workshops and symposia alone or in collaboration with other focus groups. We presented more webinars and you will find this information in the News Letter. The selection committee is already working on identifying a 2013 winner of Innovation in Nanotechnology Award, which will be presented in San Antonio later this year. So, out full and interesting life goes on and I strongly believe that this new issue of the News Letter will add more colors to this life and you will enjoy reading it. I think, you will like the section on translational nanotechnology, which opens our discussion with industry representatives who is trying to convert the results of our bench exercises in real products. Welcome and help us to make more interesting and useful issues of nanotechnology News Letter in the future. INSIDE THIS ISSUE: 2. 2012-13 GOALS AND ACCOMPLISHMENTS 3. HIGHLIGHTS OF THIS YEAR’S MEETINGS/Webinars 4. 2012 NANOTECHNOLOGY INNOVATION AWARD 5. HOT TOPIC: PROCESS AND SCALE-UP ISSUES FOR NANOMEDICINES 6. UPCOMING EVENTS AND LIST OF PROGRAMMING IDEAS Nanotechnology These figures represent fluorescence and MRI-based images of tumor cells and tumor-bearing mice showing uptake of various classes of polymeric and lipid based nanoparticles. Courtesy Centre of Pharmaceutical Biotechnology and Nanomedicine, headed by Dr. Vladimir Torchilin at Northeastern University, Boston, MA.

Transcript of NG Focus Group-Newsletter II · David Putnam, Prabhas Moghe, Chun Wang, Zheng-Rong Lu, Thomas...

Page 1: NG Focus Group-Newsletter II · David Putnam, Prabhas Moghe, Chun Wang, Zheng-Rong Lu, Thomas Anchordoguy, Richard Gemeinhart, and David ... for Biomedical Imaging and

June 2013, Issue II

2013 AAPS Nanotechnology Focus Group News-letter

1

Message From Chair

University Distinguished Professor and Director, Center of Pharmaceutical Biotechnology and Nanomedicine, School of Pharmacy, Northeastern University.

2

Dear Readers, You are keeping in hands the second issue of our New Letter. The enthusiastic team led by Shardool Jain, PhD candidate from Northeastern, has put together another set of materials relating to the activity of the Nanotechnology Focus Group of AAPS. A lot happened over the months since the first issue of News Letter was brought to you. In addition to Physical Pharmacy and Biopharmaceutics (PPB) and Formulation Design and Development (FDD) Sections, Nanotechnology FG became affiliated with Drug Discovery and Development Interface (DDDI) Section of AAPS, which still further strengthen our networking within the AAPS and improved our visibility. Group representatives took active participation in preparation of 2013 and 2014 Biotechnology Conferences and AAPS Annual Meetings, where

3

our FG will be represented by various forums, workshops and symposia alone or in collaboration with other focus groups. We presented more webinars and you will find this information in the News Letter. The selection committee is already working on identifying a 2013 winner of Innovation in Nanotechnology Award, which will be presented in San Antonio later this year. So, out full and interesting life goes on and I strongly believe that this new issue of the News Letter will add more colors to this life and you will enjoy reading it. I think, you will like the section on translational nanotechnology, which opens our discussion with industry representatives who is trying to convert the results of our bench exercises in real products. Welcome and help us to make more interesting and useful issues of nanotechnology News Letter in the future. INSIDE THIS ISSUE:

2. 2012-13 GOALS AND ACCOMPLISHMENTS 3. HIGHLIGHTS OF THIS YEAR’S MEETINGS/Webinars 4. 2012 NANOTECHNOLOGY INNOVATION AWARD 5. HOT TOPIC: PROCESS AND SCALE-UP ISSUES FOR NANOMEDICINES 6. UPCOMING EVENTS AND LIST OF PROGRAMMING IDEAS

Nanotechnology

These figures represent fluorescence and MRI-based images of tumor cells and tumor-bearing mice showing uptake of various classes of polymeric and lipid based nanoparticles. Courtesy Centre of Pharmaceutical Biotechnology and Nanomedicine, headed by Dr. Vladimir Torchilin at Northeastern University, Boston, MA.

Page 2: NG Focus Group-Newsletter II · David Putnam, Prabhas Moghe, Chun Wang, Zheng-Rong Lu, Thomas Anchordoguy, Richard Gemeinhart, and David ... for Biomedical Imaging and

Lorem Ipsum

Under the leadership of Dr. Vladimir Torchilin the main focus of the group was to conduct AAPS sponsored workshops, participate in the programming of National Biotechnology Conference and Annual Meeting, and conduct webinars to update the AAPS members and affiliated about the latest developments in the field of nanotechnology. Following is a summary of some of the major workshops/seminars that were conducted during last year:

2012 AAPS Annual Meeting, Chicago, IL Mini-Symposium “Lipidic Nano-carrier Delivery of “Big” Molecules” The symposium was focused on the use of nano-structured lipo-polymeric injectable nano-carriers for the therapeutic delivery of macromolecules such as proteins, enzymes, antibodies, peptides, genes, siRNAs, and vaccines and small molecule drugs in prevalent human diseases like cancer, and cardiovascular disorders, via describing various approaches and analysis techniques involved in the following focus topics: • Synthesis control and up-scalability of and bio-conjugation of novel nanostructured materials for therapeutic delivery; • In vitro studies on nanomaterial-drug interactions, drug release kinetics, biocompatibility and efficacy of nanomaterial-drug composites; • Targeted Nanomaterial-mediated improvement of macromolecule biodistribution, pharmacokinetics and pharmacodynamics; • Multi-functional drug carriers (targeting efficiency and feasibility studies, in vitro and in vivo) • Nanomaterial-enabled combination drug delivery (in vitro and in vivo challenges) • Localization, bio-fate/ biodistribution, and biocompatibility of nano-structured materials; • Nanotechnology for crossing biological barriers (assessment and analysis) • Stimuli triggered delivery and drug release from nanocarriers (design requirements, challenges, and strategies) • Preclinical studies on nanomaterial-supported delivery of proteins, peptides, enzymes, antibodies, genes, siRNAs, vaccines, small molecule drugs for cancers and other diseases. • Translational studies and approval challenges of nanomaterial-supported macromolecule drug delivery. • Regulatory challenges of macromolecule drug nano-formulation. 2013 National Biotechnology Meeting San Diego, CA-May 19-22, 2013 Symposium: Tuesday, May 21, 2013 (1:30 PM -4:30 PM) Nanotechnology Platform-based Biomarker Assays This symposium showcased some of the most promising and exciting nanotechnology-based case studies for interrogation of biomarker and will provide the audience with a unique opportunity to hear the experts and evaluate the promising advances in this field. List of talks/speakers as part of this seminar series are highlighted below: • Single Cell Analysis of Intracellular Content with Nano Flares: Chad Mirkin, PhD., Professor, Northwestern University. • Magneto-Nano Chips for Ultrasensitive and Multiplex Detection of Protein Biomarkers of Tumor: Shan X. Wang,

PhD., Professor, Stanford University. • Nanovelcro-Embedded Microchips that Enable Molecular and Functional Analyses of Circulating Tumor Cells

(CTCs). Hsian-Rong Tseng, PhD. Associate Professor, University of California • Nano-enabled tools for highly multiplexed detection of protein and cellular biomarkers. Rong Fan M.S., Assistant

Professor, Yale University.

Part II: 2012-13 GOALS AND

ACCOMPLISHMENTS

These figures represent Raman spectroscopic images of HeLa cells showing uptake of lipid based PEG-PE micelle (left) and TAT-peptide modified micelle (right) formulations. Courtesy Centre of Pharmaceutical Biotechnology and Nanomedicine, headed by Dr. Vladimir Torchilin at Northeastern University, Boston, MA.

AAPS nanotechnology focus group June 2013, Issue II

Page 3: NG Focus Group-Newsletter II · David Putnam, Prabhas Moghe, Chun Wang, Zheng-Rong Lu, Thomas Anchordoguy, Richard Gemeinhart, and David ... for Biomedical Imaging and

AAPS nanotechnology focus group June 2013, Issue II

Part III: Highlights of This Year’s Meetings/Seminars

1

The 10th International Nanomedicine and Drug Delivery (NanoDDS’12)

Symposium, Atlantic City, New Jersey

Co-Chairs: Professors Tamara Minko and Arash Hatefi

(Rutgers, the State University of New Jersey)

The 10th International Nanomedicine and Drug Delivery (NanoDDS’12) Symposium was originally organized to be held in Atlantic City, New Jersey on October 29-30, 2012; however, it got cancelled due to Hurricane Sandy which landed on the same day and place. Despite the circumstances, the organizers (Professors Tamara Minko and Arash Hatefi from Rutgers University) were able to reschedule the NanoDDS’12 symposium for on December 6-7, 2012 and have it held in the same place.

The overall objective of the 10th International Nanomedicine and Drug Delivery Symposium (NanoDDS’12) was to bring together a cadre of internationally recognized experts in the area of nanoscale-based drug delivery systems to discuss the challenges associated with nanomedicines. The Symposium was specifically focused on nanocarriers of different composition, architecture, size and charge, and their use for treatment and imaging. While in several other scientific conferences and meetings of well-established professional societies the advances in nanomedicine and bio-imaging research are discussed, the NanoDDS series of symposia are focused on discussing the challenges and shortcomings of the nanomedicines and elaborating on potential solutions. . The NanoDDS symposia series started in 2003 and have been established as a

2

medium size, internationally recognized symposia closely focused on aspects of nanomedicines that are related to drug delivery and imaging.

Jindrich Koopecek (University of Utah), Alexander Kabanov (University of North Carolina, Chapel Hill), and Abraham Rubenstein (Hebrew University, Jerusalem) delivered the first three of the symposium’s 26 lectures, speaking on “Novel Nanostructures and Nanomaterials.” The morning session ended with presentation on “Nanosphere-based formulations of highly hydrophobic drugs for topical delivery” by Joachim Kohn of Rutgers University. Presenters talked about design of drug-free macromolecular therapeutics, state of the art drug delivery systems, development of biomarkers for real-time diagnostics of malignancy, the challenges that nanomedicines face before translation into the clinic and future of drug delivery. Julia Ljubimova (Cedar-Sinai Medical Center, CA.), Diane Burgess (University of Connecticut), Ram Mahato (University of Tennessee), and Robert Prud’homme (Princeton University, N.J.) delivered their lectures in the second morning session that was focused on the “Nanosystems for Theranostics and Diagnostics.” Afternoon sessions were focused on the “Engineered Nanosystems and Nanomaterials” and “New Technologies in Nanomedicine and Drug Delivery.” Professors Andrew Mackay, David Putnam, Prabhas Moghe, Chun Wang, Zheng-Rong Lu, Thomas Anchordoguy, Richard Gemeinhart, and David Oupicky shared their new findings with colleagues. The symposium’s second day was mainly focused on the “Multifunctional and Multicomponent Nanosystems for Drug Delivery.” Leaders in the field as well as new investigators, Adah Almutari, Vinod Labhasetwar, Jayanth Panuam, Tatiana Bronich, Mark Tracy, and Oleh Taratula presented very interesting, original and novel research works in the session. Researchers in academia and industry from across the United States, as well as England, India, Iran, Japan, Switzerland, and Russia presented more than 60

3

posters at the symposium. Subjects included delivery of nanozymes, drugs and nucleic acids for treatment of cancer, stroke, cardiovascular, lung and other diseases using well-defined and novel nanostructures.

A special session on “NIH Grantsmanship in Nanomedicine and Nanotechnology areas” conducted by Steven Zullo of the National Institute for Biomedical Imaging and Bioengineering and Yali Fu of the National Cancer Institute attracted considerable attention of participants. James Li and Amy Rubenstein of NIH/CSR gave a brief overview of the NIH-peer review process, particularly the nanomedicine and drug delivery study sections.

The symposium was successful in bringing together new investigators and students and also well-known world-recognized scientists from academia and industry. The relatively small number of participants enabled young investigators to discuss their research with recognized specialists in the field and helped them establish network connections for further collaborations and employment. The specific objectives of the symposium were: (1) to facilitate translational research by enhancing the discussion, exchange of opinions, and establishing collaborations between academic scientists, industrial and clinical researchers; (2) to provide a forum to discuss theoretical and applied problems in nanomedicine and drug delivery; and (3) to allow students, postdoctoral researchers, and young investigators to present and discuss their research in front of world recognized scientists. The feedbacks from the participants were very positive and overall it was believed that the symposium was successful in achieving its objectives.

Page 4: NG Focus Group-Newsletter II · David Putnam, Prabhas Moghe, Chun Wang, Zheng-Rong Lu, Thomas Anchordoguy, Richard Gemeinhart, and David ... for Biomedical Imaging and

AAPS nanotechnology focus group June 2013 Issue II

Part III: Highlights of This Year’s Meetings/Webinars Contd.

The inaugural Nanomedicines Alliance Industry Symposium, Nanomedicines: Charting a Roadmap to Commercialization was held on March 6-7, 2013 at the Hilton Washington DC/Rockville Hotel. Seventy attendees representing government, academia and the pharmaceutical industry participated in the symposium, which featured plenary presentations from nanomedicine leaders and addressed five distinct sections:

· Designing Nanomedicines, which addressed recent developments in the area of design, highlighting the need for collaboration in various fields ranging from biology, chemistry, physics, and engineering.

· Preclinical Pharmacology, featuring a review of nonclinical animal studies and considerations into translating these into safe and effective human medicines.

· Chemistry, Manufacturing and Controls (CMC), which focused on case studies illustrating CMC challenges specific to nanomedicines, and potential solutions to these challenges.

· Toxicology/Absorption, Distribution, Metabolism and Excretion (ADME) addressed the evaluation of toxicological properties and ADME patterns, with focus on the immune system, stability and deposition, and vitro/in-vivo correlation.

· Clinical Studies focused on unique tools, adaptive trial designs, and other innovations that may facilitate clinical development for nanomedicines.

Importantly, representatives from government agencies and labs such as the FDA, NCL, NIH, EPA, GAO actively participated. The symposium included extensive breakout sessions in each of the focus areas with dialogue among regulators from the FDA, NCL, NIH, academia and the pharmaceutical industry. These sessions addressed practical challenges in getting nanomedicines into the market as well as possible solutions. One attendee stated, “For the first time, we had an opportunity to discuss concerns about the development of nanomedicines into the market as well as possible solutions. One attendee stated, “For the first time, we had an opportunity to discuss concerns about the development of nanomedicines with big pharma.” The symposium also featured a poster session that included 11 posters representing case studies and research in the five focus areas.

Feedback from symposium participants was overwhelmingly positive regarding the content and structure, but the overall networking opportunity and the solicitation for input on the topics were particularly well received.

Webinar Series 1. Potential of Nanotechnology in Gene Therapy. January 10, 2013 − Conducted by Dr. Leaf Huang, Eshelman

School of Pharmacy, University of North Carolina, Chapel Hill

− Moderator: Dr. Ram Mahato, University of Nebraska

The objective of this webinar was to provide basic understanding of concepts in gene therapy, discuss major barriers in developing delivery platforms for siRNA and/or miRNA, provide a road-map to effective gene delivery with emphasis on examples of non-viral based nanoparticle platform technologies.

The main topics covered in the webinar were: • Introduction to Gene

Therapy. • Roadmap of Effective Gene

Delivery. • Viral vs. Non-Viral Vectors

for Gene Therapy • Potential of siRNA/miRNA

Technology and Delivery Challenges.

• Specific Examples of Non-Viral Gene Delivery Platforms for siRNA.

2. Nanotechnology for Multi-Modality Imaging and Theranostics. February 21, 2013. − Conducted by William Phillips,

MD, The University of Texas Health Science Center at San Antonio, TX.

− Moderator: Dr. Tamer Elbayoumi, Midwestern University, AZ.

This webinar was focused on reviewing some of these multi-functional nanoparticle based platform technologies with specific examples. The webinar will be beneficial to scientists who are interested in the field of imaging and want to learn the potential of nanotechnology in this field.

The main topics covered in the webinar were: • Introduction to

Nanotechnology. • Current Issues with State-of-Art

Imaging Modalities • Potential of Nanotechnology

and Challenges in Field of Imaging.

• Specific Examples of Multi-Functional Delivery Platforms for Multi-Modality Imaging.

• Specific Examples of Multi-Functional Delivery Platforms for Theranostics.

Marc J. Wolfgang, VP Pharm Sci & Manufacturing, Cerulean Pharma, MA

Page 5: NG Focus Group-Newsletter II · David Putnam, Prabhas Moghe, Chun Wang, Zheng-Rong Lu, Thomas Anchordoguy, Richard Gemeinhart, and David ... for Biomedical Imaging and

2012 Innovation in Nanotechnology Award was presented at the Annual Meeting to Dr. Upadhyay from Department of Pharmaceutical Sciences at University of Colorado, Denver.

Dr. Arun Kumar Upadhyay graduated from a prestigious institute, National Institute of Immunology, New Delhi, India. He has expertise in areas of large scale production and biochemical analyses of therapeutic proteins, development of stable formulations, novel therapeutic protein engineering, development of protein sustained delivery systems, and design of protein targeted transcellular delivery systems. Dr. Upadhyay has designed of many superior nano- and micro- assemblies of proteins and pH shift assembly of Ad5 viral capsid to load small nanoparticles, proteins, and nucleic acids. Further, he demonstrated that surface transferrin content influences cell uptake of functionalized nanoparticles in a transferrin-receptor-mediated manner. Using the functionalized nanoparticles loaded with diclofenac, he showed that drug delivery to the back of the eye for a week, allowing once a week dosing.

The study conducted by Dr. Upadhyay which led him to win the prestigious “Innovation in Nanotechnology” award was the development of protein nanosystems having enhanced stability and activity. In this study he developed various nano- and micro-sized assemblies of alpha crystallin B (D3), a mutant chaperone protein. This protein has ability to prevent protein aggregation. He demonstrated that alpha crystallin B (D3) assemblies have unique size-dependent ability to enhance chaperone activity. He showed that nano-assemblies were superior when compared to micro-assemblies in enhancing chaperone activity. This enhanced chaperone activity of nano-assemblies was potentially contributed by altered conformation and thermal melting behavior, increase in alpha helicity, and presence of large percentage of accessible hydrophobic surface, compared to the parent molecule. The nano- assemblies are more stable than the native protein and exhibited enhanced intracellular delivery and in vivo persistence. Since therapy with chaperone proteins is limited by their low potency, poor intracellular delivery, and low in vivo persistence, the findings of this study are of great significance in translating chaperone proteins. Chaperone nano-assemblies are of value in treating protein aggregation disorders in various organs ranging from the eye to the brain. Further, the approaches and compositions for preparing nano-assemblies used in this study will likely pose no barriers for human translation.

Since this study advances design of potent chaperone nano-assemblies with enhanced stability, cellular delivery, and in vivo persistence, it can potentially be translated for therapeutic purposes in treating/preventing broad classes of protein aggregation disorder.

Part IV: 2012 Innovation in Nanotechnology Award

"The revolutionary promise of molecular nanotechnology (MNT) has become a part of society's expectations for the future. This technology will provide nanomedicine breakthroughs that could cure cancer and extend life space, bring abundance without environmental harm and provide clean sources of energy. These ideas are part of the vision that launched the field of nanotechnology." ~K. Eric Drexler

Page 6: NG Focus Group-Newsletter II · David Putnam, Prabhas Moghe, Chun Wang, Zheng-Rong Lu, Thomas Anchordoguy, Richard Gemeinhart, and David ... for Biomedical Imaging and

Part V: Hot Topic in Nanotechnology. Process and Scale-Up Issues for Nanomedicines

AAPS nanotechnology focus group June 2013 Issue II

The goal of nanomedicine development is a viable drug product that will be regulated by some government regulatory agency such as the FDA or European Medicines Agency (EMA). One of the most critical issues involved in drug development is the need of discovery personnel to identify and incorporate development practices generally assessed during scale-up and cGMP manufacture of an eventual drug product. This approach is not always the case, and can lead to significant delays in process development, scale-up, and manufacturing. In 2002, the FDA launched a new perspective for drug development and manufacturing, Pharmaceutical cGMPs for the 21st Century: A Risk-Based Approach (the Pharmaceutical cGMP initiative), which is applied to human drug, biological, and veterinary drugs with the following objectives:

• To encourage the early adoption of new technological advances by the pharmaceutical industry, • To facilitate industry application of modern quality management techniques, including implementation of quality

systems approaches, to all aspects of pharmaceutical production and quality assurance, • To encourage implementation of risk-based, Quality by Design (QbD) approaches that focus both industry and

Agency attention on critical areas, • To ensure that regulatory review and inspection policies are based on state-of-the-art pharmaceutical science, • To enhance the consistency and coordination of FDA's drug quality regulatory programs, in part, by integrating

enhanced quality systems approaches into the Agency's business processes and regulatory policies concerning review and inspection activities.

Although those individuals with experience in small and large molecule drug development would claim that scale-up issues for those products are complex and varied, and may be difficult to integrate into the FDA’s 21st century approach, the emergence of nanoparticle applications to drug development has ushered in a new level of challenges due to increased complexity and variety in mechanistic approaches, analytics, and general manufacturing practice. There are a wide variety of nanotechnology products under development such as lipid nanosystems, nanocrystals, polymeric nanoparticles, and micellar systems. These products may employ simple monomeric formulations, or may be multifunctional complex mixtures of active components and/or supportive excipients.

Post-discovery steps in the scale-up of a new drug entity can be broken into inter-reactive categories, 1) process transfer from drug discovery, 2) sub-component manufacturing (most notable multifunctional complexes), 3) analytics development and qualification, 4) formulation, 5) stability, 6) training, and 7) regulatory drivers (Table 1). This article will focus primarily on multifunctional complexes, but is the details are applicable across the entire range of nanoparticle development.

In the case of multifunctional nanomedicines, many of the ingredients used in the formulation will be unique to that product and will be manufactured just prior to, or in parallel with the final drug product by the manufacturer or a qualified vendor. An example of a multifunctional drug product would encapsulate one or more active ingredients,

David William President Blue Ocean Bio-manufacturing, Inc. Worcester, MA

Dr. Timothy P. Coleman CEO & President, Nemucore Medical Innovations Worcester, MA

Page 7: NG Focus Group-Newsletter II · David Putnam, Prabhas Moghe, Chun Wang, Zheng-Rong Lu, Thomas Anchordoguy, Richard Gemeinhart, and David ... for Biomedical Imaging and

AAPS nanotechnology focus group June 2013, Issue II

Table 1. Process Development and Scale-up steps after initial drug discovery

Process transfer

Sub-component manufacturing/purchase/evaluation, Perform bench scale to verify process steps, Modify process steps as necessary for process scale-up to specified levels, Establish baseline for process yields and recoveries, Identify potential scale-up issues during process development scale-up

runs at pre-determined scale, Modify PFDs (process flow diagrams), Develop fill operation processes and procedures.

Analytics

Evaluate or modify assay methods to compare with previous performance, Initiate development of Analytical Control Strategy, Compare PD (process development) and scale-up product to original reference standards.

Formulation Assess formulation & container closure specifications and incorporate into PFDs.

Stability Test product for stability, real time and accelerated, and compare to any previous data.

Training Provide hands-on experience for process technology and product staff (Including analytical and manufacturing staff)

Regulatory Review/institute all regulatory drivers that impact scale-up/manufacturing.

A planned indication statement for the final drug product, along with other aspects of the desired quality target product profile, should guide a development program (FDA Draft Guidance Document: Guidance for Industry and Review Staff: Target Product Profile — A Strategic Development Process Tool, March 2007). A draft target product profile should be developed as part of the process development and IND process, and should be updated throughout the development program. A generalized indication statement and other key aspects of the desired target product profile are shown below.

Selected Elements of the Desired Target Product Profile

• Indication Statement • Usage • Dosage and Administration • Dosage Forms and Strengths • Contraindications • Warnings and Precautions • Adverse Reactions • Drug Interactions • Use in Specific Populations • Overdosage • Description

• Clinical Studies • References • How Supplied/Storage and Handling • Patient Counseling Information

one or more targeting or active ligands, and/or an imaging agent. With such a complex mixture of dissimilar function ingredients, process and product characterization will be extremely challenging. Each component brings its own set of release criteria, contaminant profile, and analytical techniques, while the final drug product may not be the sum characteristics of its individual components, and may have its own unique characteristics, and product identity profile.

Page 8: NG Focus Group-Newsletter II · David Putnam, Prabhas Moghe, Chun Wang, Zheng-Rong Lu, Thomas Anchordoguy, Richard Gemeinhart, and David ... for Biomedical Imaging and

Part V: Hot Topic in Nanotechnology Contd.

AAPS nanotechnology focus group June 2013, Issue II

Raw Materials and Components ICH Q7A ICH Harmonised Tripartite Guideline, Guidance for Industry, Q7A Good Manufacturing Practice Guidance for Active Pharmaceutical ingredients, August, 2001, outlines criteria for handling and use of raw materials and components used in the production of an API (active pharmaceutical ingredient). These principles also hold for all excipients and components used in the formulation of final drug product. During technology transfer activities, a raw materials (RM) list is generated from discovery records, process description, and PFDs. The typical materials management program would consist of the following components: 1) identify raw materials and components used in the manufacturing process, 2) review regulatory requirements, 3) source raw materials, critical reagents, and components, 4) audit RM and component suppliers, if required, 5) draft RM specifications, 6) establish sampling, testing, inventory management and storage plans (ICH Q7A), 7) procure raw materials and 8) test & release raw materials and components. A safety review of raw materials should be conducted to determine issues of personnel safety, equipment suitability for the use of toxic or hard to handle raw materials, and general handling and disposal of noxious or toxic raw materials or intermediates.

Process Design/Process Characterization

The FDA considers Process Characterization as part of Process Design, the stage where process development will define the Design Space. Design Space is defined as “the multidimensional combination and interaction of input variables (e.g., material attributes) and process parameters that have been demonstrated to provide assurance of quality. Working within the design space is not considered as a change. Movement out of the design space is considered to be a change and would normally initiate a regulatory post-approval change process. Design space is subject to regulatory assessment and approval (ICH Q8 [R2]). The Process Characterization campaign will follow a QbD approach and would include the following steps (i) Refine the QTPP (Quality Target Product Profile), (ii) determine quality attributes, (iii) perform a risk analyses to identify parameters for process characterization, (iv) design and execute studies using design of experiments (DOE) and analyze results to determine the importance of the parameters as well as their role in establishing Design Space, (vi) establish a control strategy and (vii) manage product lifecycle. Figure 1 shows a QbD approach to product development.

The process is characterized by developing small-scale models, simulating commercial production scale, and defining the Design Space. QbD includes a systematic evaluation, understanding and refining of the formulation and manufacturing process by identifying, through prior knowledge, experimentation, and risk assessment, the material attributes and process parameters that can have an effect on product quality attributes (QAs). Drug Substance Critical Quality Attributes (CQA) typically includes properties related to impurities since they have a direct impact on drug safety. Impurities would include side reactions during synthesis, induced contaminants from the production environment, and raw material contaminants.

Figure 1. QbD Approach to Drug Development.

Page 9: NG Focus Group-Newsletter II · David Putnam, Prabhas Moghe, Chun Wang, Zheng-Rong Lu, Thomas Anchordoguy, Richard Gemeinhart, and David ... for Biomedical Imaging and

Part V: Hot Topic in Nanotechnology Contd.

AAPS nanotechnology focus group June 2013, Issue II

Process Characterization Risk Management and Analysis Risk management involves the identification, understanding, control, and prevention of failures that can result in manufacturing quality and product safety hazards. If any risk is judged unacceptable, it should be reduced to acceptable levels by appropriate means.

An overall risk management process involves the essential steps in Figure 2. In order to manage risk, hazards must first be identified. By evaluating the potential consequences of hazards and their likelihood, a measure of risk can be estimated.

Risk assessment, as part of risk management, consists of the identification of hazards and the analysis and evaluation of risks associated with exposure to those hazards. Quality risk assessments begin with a well-defined problem description or risk question. When the risk in question is well defined, an appropriate risk management tool and the types of information needed to address the risk question will be more readily identifiable. As an aid to clearly defining the risk(s) for risk assessment purposes, three fundamental questions are asked:

1. What might go wrong? 2. What is the likelihood (probability) it will go wrong? 3. What are the consequences (severity)?

Risk analysis is the estimation of the risk associated with the identified hazards. It is the qualitative or quantitative process of linking the likelihood of occurrence and severity of harms. In some risk management tools, the ability to detect the harm (detectability) also factors into the estimation of risk.

The QTPP table should be used to compile an initial list of CQAs, and based on their criticality, will be identified as potential materials and process parameters that could affect the CQAs. Tools typically used in the risk assessment may include the implementation of an Ishikawa or fishbone diagram, a failure mode effect analysis (FMEA), and a Pareto analysis.

A Failure Mode Effect Analysis (FMEA) is probably the most often used tool to prioritize risks and monitor the effectiveness of risk control activities. FMEA might be extended to incorporate an investigation of the degree of severity of the consequences, their respective probabilities of occurrence, and their detectability, thereby becoming a Failure Mode Effect Analysis. A FMEA can identify places where additional preventive actions might be appropriate to minimize risks. FMEA application in the pharmaceutical industry should mostly be utilized for failures and risks associated with scale-up and manufacturing processes; however, it is not limited to this application. The output of a FMEA is a relative risk “score” for each failure mode, which is used to rank the modes on a relative risk basis (ICH Q9).

Figure 2. Overall Risk Management Process

Page 10: NG Focus Group-Newsletter II · David Putnam, Prabhas Moghe, Chun Wang, Zheng-Rong Lu, Thomas Anchordoguy, Richard Gemeinhart, and David ... for Biomedical Imaging and

Part V: Hot Topic in Nanotechnology Contd.

AAPS nanotechnology focus group June 2013, Issue II

Determination of Parameter Ranges for Critical Process For each unit operation, a series of experiments should be completed using a statistically designed experimental model (e.g., DOE model such as fractional factorial). Those experiments should be used to determine which variables have a critical impact on the process performance with respect to product purity, homogeneity etc. These variables will constitute the set of parameters whose specification will be critical to reliable process performance.

Further experiments for each of the Critical Process Parameters for each of the unit operations in the process should be completed to establish proven acceptable ranges. The studies should be designed to determine the effects of excursions beyond the specified ranges on product quality. Experiments will be performed at a small scale, either using DOE approach or a systematic single variable study depending on the number of critical parameters identified for each unit operation. Studies should be done with unit operations for each ingredient for primary production/purification, and for final drug product formulation studies.

Impurity Removal Studies In establishing a validated process, it will be critical to demonstrate that all the impurities that are present in the initial material can be reliably removed with the specified process. This removal process may be a major challenge when developing a multifunctional drug product. Three categories of impurities should be addressed in any Process Characterization studies, including:

§ Process Residuals (raw materials, side-reactants). The process development activities will identify the process step(s) that remove each impurity and the capacity of the process step(s) for removal of these impurities. The latter should be demonstrated using spiking studies if applicable. Assays that can measure trace amounts of residuals will be required by regulatory guidance.

§ External Impurities (Non-process raw material induced contaminants, leachable from equipment). Quantitative data on the effectiveness of each step in removing these impurities should be developed. These are important studies, as the presence of external impurities may be considered CQAs. The limits of removal for each process step should be determined using spiking studies, if possible. Qualified assays to measure these impurities should be developed during the process optimization stage.

§ Product related impurities (Such as precipitates, degradation products, modified products). The generation and analysis of product related impurities are probably the most critical during process development and scale-up, especially when multifunctional products are being developed. Many multifunctional products are noted for their instability and leaching of active ingredients, ligands, and excipients. If impurities are present, quantitative data on the removal of these impurities at each purification step and during final formulation should be developed. The assays to measure these impurities should be qualified during the process development stage and validated during late clinical manufacture.

Process Performance Qualification (PPQ) Protocols for API and finished drug product (FDP) The eventual goal for process development and scale-up activities will be to develop a scalable and reproducible product based on PPQs for components and final drug product that will specify the manufacturing conditions, controls, testing, and expected outcomes that are essential for process validation and should contain the following (Guidance for Industry. Process validation: General Principles and Practices. 2011):

§ Manufacturing conditions, including operating parameters, processing limits, and component (raw material) inputs.

§ A plan for collecting and reviewing data. § An analytical testing plan (in-process, release, characterization) and acceptance criteria for each significant

processing step. § A detailed, extensive sampling plan describing frequency and number of samples for each unit operation.

Page 11: NG Focus Group-Newsletter II · David Putnam, Prabhas Moghe, Chun Wang, Zheng-Rong Lu, Thomas Anchordoguy, Richard Gemeinhart, and David ... for Biomedical Imaging and

Part V: Hot Topic in Nanotechnology Contd.

AAPS nanotechnology focus group June 2013, Issue II

§ A description of the statistical methods to be used in analyzing all collected data (e.g., statistical metrics defining both intra-batch and inter-batch variability).

§ Provision for addressing deviations from expected conditions and handling of nonconforming data. § Review and approval of the protocol by appropriate departments and the quality unit.

Other documents and activities required during the latter stages of Process Qualification are:

§ Cleaning validation § Stability testing § Analytical validation § Raw materials Qualification § Facility Qualification § Documentation The processes for preparation of ingredients and FDP will be locked at the end of the Process Qualification phase, and process controls will have been establish per the Product Performance Qualification design studies. At this point, criteria for the product and formula (QTPP), process description with PFDs and the rationale for each step, Critical Process Parameters and process for determining criticality to the associated Critical Quality Attributes, a description of Process Design space and methodology, container closure and packaging selection, validated test methods for safety, purity, identity, strength and quality and specifications for API, FDP and stability should be completed to support commercial manufacturing.

cGMP Documentation During process development and scale-up activities, attention should be paid to the development of documentation required to support cGMP production. Production records should be divided into several sections that provide instructions for various unit operations. For example, production records will consist of separate batch records for synthesis, recovery and purification of components, as well as batch records for formulation, filling operations, and labeling and packaging. In addition, other process documents may include any associated protocols that support the manufacturing process, i.e., formulation solution preparation, equipment SOPs, etc. Documentation examples may include the following:

§ Process Description § Batch Production Records § PFDs § Product/RM Specifications § Analytical Documentation

Overall Regulatory Guidance for Nanomedicines Despite the advances in nanomaterial application in disease diagnosis and drug delivery, a significant amount of work still needs to be done in terms of characterizing nanomedicine safety and long term effects on biological systems. At this time, there are no standardized regulatory guidelines for nanomedicines, and additionally, there is currently no standardized nomenclature or definitions for nanotechnology within the US Food and Drug Administration (FDA) or other world regulatory agencies.

Current FDA Regulations: Without specific regulatory guidance for nanomedicine, the U.S. FDA has approached product approval on a case-by-case basis. Currently, all nanomedicines go through the FDA’s traditional regulatory pathway within the Center for Drug Evaluation and Research (CDER) or Center for Devices and Radiological Health (CDRH). This pathway includes the following general requirements prior to approval.

Page 12: NG Focus Group-Newsletter II · David Putnam, Prabhas Moghe, Chun Wang, Zheng-Rong Lu, Thomas Anchordoguy, Richard Gemeinhart, and David ... for Biomedical Imaging and

Part V: Hot Topic in Nanotechnology Contd.

AAPS nanotechnology focus group June 2013, Issue II

1. CDER reviews applications for new or generic drugs.

II. Prior to clinical testing, laboratory and animal testing is performed to determine pharmacokinetic and pharmaco-dynamic attributes of the drug to determine a likely safety and toxicology profile in humans.

III. Clinical trials are performed in stages to determine if the drug is safe in healthy, then sick patients,

and whether it provides a significant health benefit.

IV. A team of FDA physicians, chemists, toxicologists, pharmacologists, and other pertinent scientists evaluates clinical data, and if safety and efficacy are established, the drug is approved for marketing.

Bridging the Gap between Discovery and Commercial Manufacturing Nanomedicine translation faces substantial challenges related to managing the complex data streams emerging from lab bench work, from process development work, and from preclinical studies, all with important attributes required for drafting a Target Product Profile (TPP). The critical information developed during these activities is required to navigate a complex regulatory environment. Without effective data capture solutions and subsequent translation of large quantities of data into shared information, it will be "challenging" to coordinate the bench level process with scale-up process development, risk management and regulatory compliance. Nemucore, Inc. is currently developing a software package designed to assist academics or early discovery bench scientists in overcoming this translational bottleneck for nanomedicines by consolidating existing drug development best practices into a single software package for use as a guide to accelerate nanomedicine development.

Nanolytics is a knowledge management system for information pertinent to development of a TPP, processes development plans, validation plans and risk management assessment needed to support effective nanomedicine translation. Nanolytics allows academic investigators early in research to contextualize the movement of a nanomedicine to the clinic. Unlike either small molecule or biologic development, the creation of nanomedicines, which are complex molecular entities, is very process and design intensive. An early-stage conceptual process has already demonstrated value of an informatics approach to identify barriers (use of equipment not compatible with scale-up) and risks (regulatory, material, COGS, etc.) to translating these discovery nanomedicines to the clinic. Nanolytics software consists of knowledge suites designed to capture as nanomedicine development knowledge from the earliest stages. Early knowledge capture should mitigate cost and reduce time of development of scale-up processes, lower barriers to clinical development for nanomedicines, and leverage research dollars more effectively. Nanolytics allows for the input of key information from initial research that can later be used to achieve pilot scale production of the target nanomedicine. As is always the case, better information, begets more realistic product development plans. The development of information "outside" of the typical areas of focus of a nanomedicine researcher will reduce risk and clarify efforts in translating nanomedicines from bench to bedside.

In summary, process development and scale-up processes should meet goals normally encountered in traditional manufacturing processes. Very early attention to process development, process scalability, equipment, analytical procedures, and regulatory requirements should all be part of the drug development process from discovery to commercial manufacturing. This may be a difficult challenge for nanomedicine development, including discovery, process development, scale-up, and regulatory consideration as it is in a nascent stage of development with few standards and guidelines. Nanomedicines may require even more diligence during the drug development process due to their potentially unique properties, intrinsic complexity, and mechanistic qualities (such as intracellular activity, PK/PD properties, or blood/brain barrier properties). On behalf of Dr. Torchilin, we would like to sincerely thank Dr. Tim Coleman and David William for their valuable thoughts on the important topic surrounding nanomedicine and contribution towards developing our annual newsletter.

Page 13: NG Focus Group-Newsletter II · David Putnam, Prabhas Moghe, Chun Wang, Zheng-Rong Lu, Thomas Anchordoguy, Richard Gemeinhart, and David ... for Biomedical Imaging and

Part VI: Upcoming Events and Programming Ideas

- Nanotechnology in Dermal and Transdermal Drug Delivery (with other FGs) - Nanotechnology risk assessment (with other FGs) - Pulmonary delivery using microparticles vs. nanoparticles - Emerging lipoprotein and lipid micellar nanoparticles for siRNA delivery - Quality control of nanoparticulate drugs - Design and testing in nanomedicine - Academia-technology interaction in nanomedicine - Challenges in manufacture nanomedicines - Nanoparticles for targeting stem cells - Nanosystems for diagnostic imaging - Polymer and lipid nanoparticles in ocular delivery - Biodistribution and PK of functionalized nanoparticles - Scaling up Nano-formulations Please note that some of these programming ideas will be used for either conducting mini symposia or webinars. Our group encourages interested speakers that are actively conducting research in any of the above-mentioned fields to connect with Dr. Torchilin in order to sign up for a webinar. In addition, AAPS member community is highly encouraged to suggest new programming ideas to the steering committee members of nanotechnology focus group. Current Leadership Profile: • Chair: Vladimir Torchilin, Ph.D, Northeastern University, School of

Pharmacy • Chair-Elect: Ram Mahato, Ph.D, University of Nebraska Medical Center,

College of Pharmacy • Marcel Bally, PhD, British Columbia, Cancer Agency, Vancouver, BC,

Canada • Ben Boyd, PhD, Victoria College of Pharmacy, Monash University, Australia • Tamer Elbayoumi, Ph.D., Midwestern University, Glendale, AZ • Joseph A. Fix, PhD, FIX Pharma Consulting, Lawrence, KS • Lee Jia, PhD, NCI/NIH, Rockville, MD • David Lechuga, PhD, Pearl Therapeutics, Redwood City, CA • Tamara Minko, Ph.D., Rutgers University, NJ • Russell J. Mumper, PhD, UNC, School of Pharmacy, Chapel Hill, NC • Rakhi Shah, PhD, FDA/CDER/DPQR, Silver Spring, MD • Shahnam Sharareh, PharmD, RAC, Attorney at Law, Fox Rothschild LLP, Lawrenceville, NJ • Marc J. Wolfgang, Cerulean Pharma, Boston, MA We would also like to thank our steering committee members for their feedback and continued support with regards to the programming ideas and planning of the activities for Nanotechnology Focus Group.

AAPS 2013 Annual Meeting and Exposition San Antonio, TX-Nov. 10-14, 2013

NG Focus is group is organizing following seminar at the annual meeting:

1. Mini-Symposium “Lipid-based systems for siRNA delivery” (Dr. Torchilin) 2. Short course “Quality control aspects of nanoparticulate Drugs” (Dr. Constantinides and Torchilin)

List of Programming Ideas:

Shardool Jain is a Doctoral candidate in Department of Pharmaceutical Sciences at Northeastern University. He is currently working under the guidance of Dr. Mansoor Amiji. The title of his thesis project is “Macrophage-Targeted Alginate-Based Non-Viral Gene Delivery System for Anti-Inflammatory Gene Therapy in Treatment of Rheumatoid Arthritis”.

This issue of news-letter was developed by him and is an active student member of the American Association of Pharmaceutical Scientists Nanotechnology Focus Group.

AAPS nanotechnology focus group June 2013, Issue II