establishing cgmp manufacturing capability for … cgmp manufacturing capability for phase 1...

24
Establishing cGMP Manufacturing Capability for Phase 1 Sterile, Dispersed, Injectable Dosage Forms Donald Pennino, Ph.D, VP, QA/ARD & Jingjun Huang, Ph.D., CEO Ascendia Pharmaceuticals LLC

Upload: ngothu

Post on 06-May-2018

272 views

Category:

Documents


2 download

TRANSCRIPT

Establishing cGMP Manufacturing Capability

for Phase 1 Sterile, Dispersed, Injectable

Dosage Forms

Donald Pennino, Ph.D, VP, QA/ARD &

Jingjun Huang, Ph.D., CEO

Ascendia Pharmaceuticals LLC

Establishing cGMP Manufacturing Capability for Phase 1 Sterile, Dispersed, Injectable Dosage Forms

1. Introduction

As a product transitions from pre-clinical development to

a clinical development phase, the manufacturing process

takes on a much greater role in the overall success of the

project. This transition is particularly difficult for emerging

pharmaceutical companies whose expertise typically lies in

the biology and chemistry of how their drug interacts with

targets in the body, and less on the engineering, regulatory

and quality aspects of manufacturing the drug product. This

critical milestone is made even more challenging when the

drug product is intended to be a sterile, injectable dosage

form as the manufacturing and quality requirements can

be overwhelming for a small company. Most small pharma

companies turn to Contract Development and Manufacturing

Organizations (CDMOs) to outsource this activity, as the cost

to establish the capability internally often does not merit

the investment for an early-stage product.

Many CDMOs do not provide manufacturing services for

injectable products, and those that do often have facilities

suitable for large scale production. Since early-stage

products are almost always manufactured in small batches,

there is a market need for CDMOs with the flexibility to

provide manufacturing services for Phase I sterile dosage

forms. For example, in some cases, it may be advantageous

for a CDMO to establish a GMP area within a “laboratory

setting” for the manufacture of drug product in early

development. The rationale for this approach is to avoid the

significant investment in setting up a dedicated facility and

to create simpler, more flexible systems that meet GMP

requirements, but are tailored for the specific activity

envisioned. As long as the appropriate GMP controls

are maintained, especially as related to operator safety,

cleaning, and prevention of cross-contamination, there is

no compliance barrier to using “lab-type” facilities for the

manufacture of early phase clinical batches. This article

describes the considerations for establishing this capability,

and focuses on dosage forms that are sterile and dispersible.

Page 2

Production of the

first clinical trial

materials for a

new pharmaceutical

dosage form is a

significant milestone

event in the

development of

a pharmaceutical

product.

Establishing cGMP Manufacturing Capability for Phase 1 Sterile, Dispersed, Injectable Dosage FormsEstablishing cGMP Manufacturing Capability for Phase 1 Sterile, Dispersed, Injectable Dosage Forms

2. Regulatory Landscape for Phase 1 Dosage Forms

On September 15, 2008 the FDA made effective an

amended rule that applies to small-molecule drugs and

biologics, including vaccines and gene therapy products.

The note in the Federal Register of 15 July 2008 (Volume

73, No. 136) announced an adaptation of 21 CFR 210 and 211:

investigational medicinal products solely intended for use

in Phase 1 are to be exempted from complying with the

“final rule” under FD&C Act 505(i) (21 U.S.C 355(i)). The

text stresses that the cGMP requirements of 21 CFR 211

are applicable only to Phase 2 and Phase 3 drugs (note,

however, the exemption does not apply once the

investigational drug has been made available for use by

or for the sponsor in a Phase 2, a Phase 3 trial, or if the

drug has been lawfully marketed).

“FDA’s position is that the United States’ [GMP]

regulations were written primarily to address commercial

manufacturing and do not consider the differences

between early clinical supply manufacture and

commercial manufacture,” the agency says.

For example, the requirements for a fully validated

manufacturing process, rotation of stock for drug product

containers, repackaging and relabeling of drugs and

separate packaging and production areas need not apply

to investigational drug products made for use in Phase 1

trials, the agency says. This makes for solid rationale, as

the typical batch size needed for Phase 1 clinical trials is

typically much smaller in comparison to Phase 3 and

commercial scale batches, and hence the critical controls

needed for Phase I should focus on safety of manufacture

rather than qualification of processes at this point of drug

development (note, however, the FDA did emphasize the

importance of meeting the “statutory GMP requirements”

of the Food and Drug Act ” 501 (a)2(B) which direct 21 CFR

Parts 210 and 211).

Page 3

Establishing cGMP Manufacturing Capability for Phase 1 Sterile, Dispersed, Injectable Dosage Forms

In connection with the final rule on Phase I drug GMPs, the

FDA issued a guidance recommending approaches to satisfy

statutory GMP requirements for such drugs:

“During product development, the quality and safety of

Phase I investigational drugs are maintained, in part, by

having appropriate [quality control (QC)] procedures in

effect,” the guidance states. “Using established or

standardized QC procedures and following appropriate

cGMP will also facilitate the manufacture of equivalent or

comparable IND product for future clinical trials as needed.”

While the basic requirements to reduce or eliminate

contamination that would cause adulteration in

non-sterile drug products are demanding, the standards

for aseptic manufacturing of medicinal drug products are

even more stringent. The pharmaceutical product must be

non-pyrogenic, in addition to a strict sterility requirement.

Medicinal drug products that do not meet the sterility and

non-pyrogenicity requirements can otherwise cause severe

harm or a life-threatening health risk to the patient. Hence,

these attributes are of utmost importance and concern

during Phase 1 manufacture. Since the injectable dosage

form must be sterile, the drug product can be terminally

sterilized in its packaging, or manufactured aseptically.

Aseptic Manufacturing RegulationsIn Europe, aseptic manufacturing of sterile products is still

seen as a last resort which is only acceptable if all methods

of terminal sterilization in the final sealed container have

be excluded. Such being not feasible or applicable, for

example when the drug substance is thermally labile, the

EU guidelines require the sterilization in the final container

closure system whenever possible. Only the stability of the

drug substance is considered, but not the container closure

system. The European Pharmacopoeia (EP) prioritizes the

terminal sterilization of the final container in manufacturing

sterile drug products. The “EU Guidelines to GMP for

Medicinal Products for Human and Veterinary Use, Annex

1, Rev. 2008, Manufacture of Sterile Products” compiles the

recommended procedures for sterile products and includes

the aspects of aseptic manufacturing.

Page 4

Establishing cGMP Manufacturing Capability for Phase 1 Sterile, Dispersed, Injectable Dosage FormsEstablishing cGMP Manufacturing Capability for Phase 1 Sterile, Dispersed, Injectable Dosage Forms

In the USA, the FDA’s 2004 publication “Guidance for

Industry Sterile Drug Products Produced by Aseptic

Processing” describes the expectations of the FDA for the

validation of aseptic processing in a more detailed manner.

This guidance updates the 1987 guidance primarily with

respect to personnel qualification, cleanroom design and

isolators, air supply system, integrity of container closure

systems, process design, quality control, environmental

monitoring, and review of production records. The use of

isolators for aseptic processing is also discussed. According

to the latest guidance, acceptance criteria for the

evaluation of media fill states that each contaminated unit

should be examined independent of the number of filled

units. The microbial environmental monitoring (more

frequency in testing) is accorded more importance to get

greater quality assurance. Table 1 lists an overview of

regulatory guidelines for aseptic processing.

Table 1. Overview of International Requirements, Guidelines

and Recommendations for Aseptic Manufacturing

Provided By Requirement / Guideline / Recommendation

PH. Eur, 5th ed., ch. 5.1.1, 20052

Methods of Preparation of Sterile Products

USP <1211>, 20165 Sterilization and Sterility Assurance of Compendial Articles (Manufacturing of Sterile Drug Products)

EudraLex4 EU Guidelines to GMP for Medicinal Products for Human and Veterinary Use, Annex 1, Rev 2008, Manufacture of Sterile Medicinal Products, 2003

ISO 13408-1 (2015)7

Aseptic Processing of Health Care Products Part 1: General Requirements

ISO 14644-1 (2015)8

Cleanrooms and Associated Controlled Environments. Part 1 (Specification for particles in air in clean rooms)

PIC/S (Pharm. Inspection Co-Operation Scheme)

Recommendation on the Validation of Aseptic Processes (2011)

PDA Technical Report No 13 rev. (2014) 16

Fundamentals of an Environmental Monitoring Program

PDA Technical Report No 22 rev. (2011) 14

Process Simulation Testing for Aseptically Filled Products

PDA Technical Report No 28 rev. (2006) 15

Process Simulation Testing for Sterile Bulk Pharmaceutical Chemicals

PDA Technical Report No 26 rev. (2008) 17

Sterilizing Filtration of Liquids

US FDA, CDER, revised 2004

Guidance for Industry, Sterile Drug Products Produced By Aseptic Processing. Validation with Media Fill.

USP <1116>, 201411 Microbiological Monitoring of Clean Rooms and Other Controlled Environment

Page 5

Establishing cGMP Manufacturing Capability for Phase 1 Sterile, Dispersed, Injectable Dosage Forms

Page 6

3. Sterile, Dispersable Dosage Forms

SuspensionsSome drugs are insoluble in all acceptable media and

must, therefore, for parenteral use, be administered as a

suspension. One advantage is that drugs in suspension are

often chemically more stable than in solution; however, their

primary disadvantage is physical stability; i.e., that they tend

to settle over time leading to a lack of uniformity of dose.

Issues with settling can be minimized by careful formulation

and by shaking the suspension before each dose is

delivered. Physical stability in suspensions is controlled by

(1) the addition of flocculating agents to enhance particle

“dispersability” and (2) the addition of viscosity enhancers

to reduce the sedimentation rate in the flocculated

suspension. Flocculating agents are electrolytes which carry

an electrical charge opposite that of the net zeta potential

of the suspended particles. The addition of the flocculating

agent, at some critical concentration, negates the surface

charge on the suspended particles and allows the formation

of floccules, or clusters of particles, that are held loosely

together by weak van der Waals forces. Since the particles

are linked together only loosely, they will not cake and may

be easily re-dispersed by shaking the suspension. Floccules

have approximately the same size particles; therefore a

clear boundary is seen when the particles settle. Viscosity

enhancers are typically hydrocolloids (natural, semisynthetic,

or synthetic) or clays use in a concentration range from 0.5%

to 5%, but the target viscosity will depend on the suspended

particle’s tendency to settle.

A couple methods are used to prepare parenteral

suspensions. First, aseptically combining sterile powder

and vehicle involves aseptically dispersing the sterile, milled

active ingredient(s) into a sterile vehicle system (solvent plus

necessary excipients); aseptically milling the resulting

suspension as required, and aseptically filling the milled

suspension into suitable containers. For example, this

process is used for preparation of parenteral procaine

penicillin G suspension. Or, second, in-situ crystal formation

Establishing cGMP Manufacturing Capability for Phase 1 Sterile, Dispersed, Injectable Dosage FormsEstablishing cGMP Manufacturing Capability for Phase 1 Sterile, Dispersed, Injectable Dosage Forms

Page 7

by combining sterile solutions. In this method active

ingredient(s) are solubilized in a suitable solvent system,

a sterile vehicle system or counter solvent is added that

causes the active ingredient to crystallize, the organic

solvent is aseptically removed, the resulting suspension is

aseptically milled as necessary, and then filled into suitable

containers. For example, this process is used for testosterone

and insulin parenteral suspensions.

Nano-ParticlesA drug’s low solubility often presents a serious challenge to

developing bioavailable dosage forms. This challenge can be

exacerbated for drugs with chemical stability issues when

solubility-enhancing approaches utilize excipients that

are incompatible with the drug substance. To overcome

these challenges many technologies have been developed

including particle size reduction to nanometer-size drug

crystals with greater surface area for dissolution, production

of amorphous solid dispersions for reducing the energy

required for dissolution, and lipid-based drug delivery

systems for dissolving a hydrophilic drug in either a lipid

or oil phase.

Nano-EmulsionsOil-in-water emulsions, which are comprised of oil

droplets dispersed in an aqueous continuous phase, can

provide unique solutions for overcoming drug solubility and

stability problems; for example, Diprivan® (propofol), an

injectable anesthetic, is an nano-emulsion.

Emulsions can be characterized as macro, micro or nano.

Macro-emulsions are typically opaque in appearance, since

the average particle size of the hydrophobic droplet in a

macro-emulsion is typically > 500 nm and thus scatters light.

Micro-emulsions and nano-emulsions are obtained when the

size of the droplet is typically in the range of 50-500 nm. In

addition, emulsions in this size range can appear translucent

or optically clear if the average oil droplet size is < 100 nm,

as droplets in that size range no longer scatter light.

Sterile, dispersible

dosage forms

include suspensions,

nano-particles and

nano-emulsions -

these last two

formulation options

are being increasingly

utilized due to their

ability to improve

bioavailability issues

with poorly

soluble drugs.

Establishing cGMP Manufacturing Capability for Phase 1 Sterile, Dispersed, Injectable Dosage Forms

The distinction between micro- and nano-emulsions

relates to their thermodynamic stability. Micro-emulsions

are thermodynamically stable due to the use of sufficient

co-solvents and co-surfactants to prevent Ostwald ripening -

essentially the coalescence of the droplets into larger

particles. Ostwald ripening is the most frequent physical

instability mechanism, although gravitational separation can

also occur with larger particles. Nano-emulsions contain

much less of the stabilizing co-solvents and co-surfactants,

and as such are meta-stable and more susceptible to

Ostwald ripening. In addition, nano-emulsions require

greater kinetic formation energy, and are usually prepared

using high-pressure homogenization or ultrasonic

generators. Because of the undesirable side-effects

caused by many solvents and surfactants, micro-emulsions

are disadvantageous compared to nano-emulsions. In

order to achieve physically stable nano-emulsions, long

chain triglyceride oils are sometimes employed, but

typically require the use of organic co-solvents or toxic

co-surfactants (e.g., Cremaphor). The addition of co-solvents

and co-surfactants significantly reduces the safety and

tolerability profile of the pharmaceutical formulation. These

excipients may not be suitable for pediatric administration,

may cause injection site pain and irritation, and are

becoming less acceptable in general for use in

pharmaceutical formulations.

Page 8

4. Facility Size & Manufacturing Space Considerations

Matching the product to an appropriate facility size is

important. Facility infrastructure typically increases along

with facility size, and facility size increases with the scale

of the pharmaceutical project. The development phase of

the drug will dictate the capacity requirements of the

formulation and fill. For larger, later stage production

activities the maximum capacity of the facility is critical to

ensure success, but this is not a critical factor for early-stage

development projects. It is important to evaluate the

product’s requirements and determine the best fit.

A “bigger is better”

philosophy is not

always true in drug

development and

processing. In early

phase trials the quantity

of the drug substance

available is often very

small. Matching the

equipment scale and

material handling

expertise with the

product is essential

in order to ensure a

successful, cost-effective

outcome.

Establishing cGMP Manufacturing Capability for Phase 1 Sterile, Dispersed, Injectable Dosage FormsEstablishing cGMP Manufacturing Capability for Phase 1 Sterile, Dispersed, Injectable Dosage Forms

Page 9

The ideal CDMO is one that can grow with a product’s

success, but this is difficult — if not impossible — to find.

Pure CMOs that manufacture high volume commercial

products typically lack the equipment and personnel to

manage a developing product that requires low volume,

and flexibility in scheduling. Companies that specialize in

small volume early stage products have staff experienced

in rapid small-scale manufacturing campaigns. A smaller

support staff generally has greater flexibility with regard to

changes and timing. The lead time for changes at a smaller

CDMO should be less than for a CMO that is use to filling lots

greater then 100,000 units per day. Although larger CMOs

have much greater capacity, they tend to be more rigid and

generally have defined systems in place that are not easily

changed. Scheduling is done well in advance (the lead time

for scheduling or bringing a product in can be six months

to a year) so the lead time for changes can be a factor.

Evaluating the structure that you require for your stage

of production is an important aspect in choosing the

CDMO that will meet your current and potential

future requirements.

5. Equipment - Both Process and Cleanroom

Cleanrooms are used in practically every industry where

small particles can adversely affect the manufacturing

process. A cleanroom is any given contained space where

provisions are made to reduce particulate contamination

and control other environmental parameters such as

temperature, humidity and pressure. The key component is

the High Efficiency Particulate Air (HEPA) filter that is used

to trap particles that are 0.3 micron and larger in size. All

of the air delivered to a cleanroom passes through HEPA

filters, and in some cases where stringent cleanliness

performance is necessary, Ultra Low Particulate Air (ULPA)

filters are used.

Establishing cGMP Manufacturing Capability for Phase 1 Sterile, Dispersed, Injectable Dosage Forms

Page 10

Cleanrooms are classified by how clean the air is. In Federal

Standard 209 (A to D) of the USA, the number of particles

equal to and greater than 0.5mm is measured in one cubic

foot of air, and this count is used to classify the cleanroom.

This metric nomenclature is also accepted in the most recent

209E version of the USA Standard. The newer standard

is TC 209 from the International Standards Organization

(ISO 14644-1). Large numbers like “class 100” or “class 1000”

refer to the 209E Standard; the standard also allows

interpolation, so it is possible to describe e.g. “class 2000.”

Cleanrooms classified using single digit numbers refer to

ISO 14644-1 standards, which specify the decimal logarithm

of the number of particles 0.1 µm or larger permitted per

cubic meter of air. So, for example, an ISO class 5 cleanroom

has at most 105 = 100,000 particles per m3. Both FS 209E

and ISO 14644-1 assume log-log relationships between

particle size and particle concentration. For that reason,

there is no such thing as zero particle concentration.

Ordinary room air is approximately class 1,000,000 or ISO 9.

Personnel selected to work in cleanrooms undergo extensive

training in contamination control theory. They enter and

exit the cleanroom through airlocks, air showers and/or

gowning rooms, and they must wear special clothing

designed to trap contaminants that are naturally generated

by skin and the body. Since Phase 1 batches usually are

small scale, one popular alternative of CDMOs is to utilize

compounding isolators for sterile manufacture. Isolators

consist of a decontaminated unit, supplied with class 100 or

higher air quality that provides uncompromised, continuous

isolation of its interior from the external environment (e.g.,

surrounding clean room and personnel).

Establishing cGMP Manufacturing Capability for Phase 1 Sterile, Dispersed, Injectable Dosage FormsEstablishing cGMP Manufacturing Capability for Phase 1 Sterile, Dispersed, Injectable Dosage Forms

Page 11

An isolator is defined as an ISO 5 enclosure if it meets the

following criteria:

• uses rapid transfer ports or another type of

decontaminated, high-integrity interface to transfer

compounding materials into the isolator;

• uses an automatic sporicidal

decontamination system;

• constantly maintains a significant overpressure

relative to the surrounding environment; and

• the manufacturer provides documentation verifying

that the isolator can maintain ISO 5 at all times.

Any Compounding Aseptic Isolator (CAI) that does not meet

all of the isolator criteria would be classified as a restricted

access barrier system (RABS). A RABS is an ISO 5 enclosure

that provides a physical separation from the compounding

area through the use of glove ports, but the openings for

transferring materials would not provide the same level of

protection as an isolator. In addition, the RABS is cleaned

and decontaminated manually.

Aseptic Manufacturing ConsiderationsAseptic manufacturing consists of a lot of single working

steps. But the whole process is only as good as the worst

single step. To achieve the aim of a sterile product, several

aspects have to be considered and have to be separately

validated. In the end, process simulation with media fill is the

key validation measure and allows the final evaluation of the

appropriateness of the whole process. It is state-of-the-art

to produce medicinal products under aseptic controlled

conditions. This control requires monitoring of the

environment. The design of the monitoring (frequency,

number of sampling sites, method of sampling, procedure

in regard of deviations etc.) is not specifically mandated;

however, the common aim is to recognize any deviation

of the validated state.

The necessity of monitoring the environment as a

key element of a quality assurance program is widely

accepted. Air, surfaces and personnel are all identified as

contamination risk sources for the environment. To come

Aseptic

processing using

isolator systems

minimizes the extent

of personnel

involvement and

separates the

external cleanroom

environment from

the aseptic

processing line.

A very high integrity

can be achieved in a

well-designed unit.

Establishing cGMP Manufacturing Capability for Phase 1 Sterile, Dispersed, Injectable Dosage Forms

Page 12

to reasonable limits, the rooms of the production areas have

firstly to be classified depending on the production step.

Limits of air, surfaces and personnel are proposed under

consideration of the official recommendations. There are

separate requirements for non-viable air particles, and for

viable organisms, and the time when the measurements are

performed (either at rest, or in operation). The differences

determine the nomenclature for clean rooms. Both

personnel and material flow should be optimized to prevent

unnecessary activities that could increase the potential for

introducing contaminants to exposed product, container

closures or the surrounding environment. Air (including

purified air) is a main source for contamination. Per EU GMP,

viable airborne particles have to be identified and regarded

in batch release. Surfaces which have immediate contact

with the product are highly critical. Indirect transfer of

particles from surfaces via air must also be taken into

account. The design of the facility (smooth surfaces without

unevenness and tears) is important to avoid contamination

and support the success of sanitization procedures.

6. Quality Control, Approach & Audits

Although quality is the responsibility of all personnel

involved in manufacturing, it is highly recommended that

individual(s) who are assigned to perform QC functions are

independent of manufacturing responsibilities, especially for

the cumulative review and release of phase 1 investigational

drug batches.

The CDMO engaged in the manufacture of phase 1

investigational drugs should follow written manufacturing

and process control procedures that provide for the

following records:

• A record of manufacturing data that details the

materials, equipment, procedures used, and any

problems encountered during manufacturing.

Production records should be sufficient to replicate

Good quality systems

produce quality

products. For that to

happen, the operators

must be well-trained,

and the proper

personnel must review

the manufacturing

documentation. Every

manufacturer should

establish a written plan

that describes the role

of and responsibilities

for the QC function.

Establishing cGMP Manufacturing Capability for Phase 1 Sterile, Dispersed, Injectable Dosage FormsEstablishing cGMP Manufacturing Capability for Phase 1 Sterile, Dispersed, Injectable Dosage Forms

Page 13

the manufacturing process. Similarly, if the

manufacture of a phase 1 investigational drug batch

is initiated but not completed, the record must

include an explanation of why manufacturing

was terminated;

• A record of changes in procedures and processes

used for subsequent batches along with the

rationale for any changes; and

• A record of the laboratory (quality control and

microbiological) that have been implemented

(including written procedures) for the production

of sterile-processed phase 1 investigational drugs.

In the early stages of drug development, processing

parameters will be adjusted to meet efficiency targets

better and/or overcome processing hurdles. The CDMO

making these adjustments should have a formal change

control system that allows the client to present this

documentation to the FDA (or other agency) during

later stage filings.

Proper QC documentation also includes a Quality

Agreement. The quality agreement should define

expectations between the CDMO and the sponsor

to review and approve documents, and how they will

communicate with each other, both verbally and in writing.

It also should describe how changes may be made to

standard operating procedures, manufacturing records,

specifications, laboratory records, validation documentation,

investigation records, annual reports, and other documents

related to products or services provided by the contract

facility. The quality agreement should also define owners’

and contract facilities’ roles in making and maintaining

original documents or true copies in accordance with cGMP.

Establishing cGMP Manufacturing Capability for Phase 1 Sterile, Dispersed, Injectable Dosage Forms

Page 14

It should explain how those records will be made readily

available for inspection. The quality agreement also should

indicate that electronic records will be stored in accordance

with cGMP and will be immediately retrievable during the

required record-keeping time frames established in

applicable regulations.

Laboratory ControlsLaboratory tests used in manufacturing (e.g., testing of

materials, in-process material, packaging, drug product)

should be scientifically sound (e.g., specific, sensitive, and

accurate), suitable and reliable for the specified purpose.

Tests must be performed under controlled conditions

and follow written procedures describing the testing

methodology. Records of all test results, procedures,

and changes in procedures, must be maintained. The main

purpose of laboratory testing of a Phase 1 investigational

drug is to evaluate quality attributes including those that

define its identity, strength, potency, and purity, as

appropriate. Specified attributes should be monitored,

and acceptance criteria applied appropriately. For known

safety-related concerns, specifications should be established

and met. For some Phase 1 investigational drug attributes, all

relevant acceptance criteria may not be known at this stage

of development as this information will be reviewed in the

IND submission.

To ensure reliability of test results, calibration and

maintenance of laboratory equipment at appropriate

intervals according to established written procedures is

required. Personnel verify that the equipment is in good

working condition when samples are analyzed (e.g., system

suitability). A representative sample from each batch of

Phase 1 investigational drug should be retained. Retention

of both the API and Phase 1 investigational drug in

containers used in the clinical trials is essential. The sample

should consist of a quantity adequate to perform additional

testing or investigation if required at a later date (e.g., twice

the quantity necessary to conduct release testing, excluding

testing for pyrogenicity and sterility). Storage and

retention the samples for at least two years following

Establishing cGMP Manufacturing Capability for Phase 1 Sterile, Dispersed, Injectable Dosage FormsEstablishing cGMP Manufacturing Capability for Phase 1 Sterile, Dispersed, Injectable Dosage Forms

Page 15

clinical trial termination, or withdrawal of the IND application

is recommended.

Finally, initiation of a stability study using representative

samples of the phase 1 investigational drug to monitor the

stability and quality of the phase 1 investigational drug

during the clinical trial (i.e., date of manufacture through

date of last administration) should be performed under ICH

temperature, humidity and light storage conditions.

AuditsA drug sponsor should always visit the CDMO’s site during

the evaluation process. This visit gives the drug developer a

good overview of how the CDMO works. Touring the facility

shows if it is a clean and functioning facility. Evaluate

whether the staff grasps the scope of your project, and

determine whether the scientists have familiarity with the

product type. Drawing on a CDMO’s experience can save

time and potentially deliver a better outcome. For instance, a

sponsor may believe that filling their product in a multi-dose

vial is the best administration method for a clinical setting.

However, this practice could lead to errors in dosing, loss of

extremely scarce product, and potentially determining the

path forward for container stability. In the early stages

of drug development, processing parameters will be

adjusted to meet efficiency targets better and/or overcome

processing hurdles. The CDMO making these adjustments

should have a formal change control system that allows the

client to present this documentation to the FDA (or other

agency) during later stage filings. After a successful site

visit, the next step is to conduct an in-depth audit.

During the audit you will review documentation systems

and discuss the project in more depth. All information and

discussions should be viewed from a quality standpoint.

First, learn about the company’s history, size, services,

financial stability, future plans for growth and technological

innovations. Then determine the training of personnel and

the expertise level of the staff. Find out about the Quality

Assurance and Quality Control systems, manuals, reviews

and methodology; also determine certifications, document

management, procedures and problem solution systems,

Establishing cGMP Manufacturing Capability for Phase 1 Sterile, Dispersed, Injectable Dosage Forms

Page 16

and equipment maintenance and calibrations. Also, look

at measurements/metrics for monitoring and controls,

deviations (the number and significance of them), technical

transfer controls, capabilities, test methods and validations,

material controls and inspections, supplier and material

qualifications, purchasing controls, and laboratory controls.

Determine the GMP compliance history, and SOP (Standard

Operating Procedures) records. From this review a drug

developer will be able to determine if a CDMO has the

technological knowledge, compliance record, and experience

to provide solutions to problems and be able to complete

documentation in a timely fashion.

7. Personnel & Training

Double gloves are often used in industrial practice, as a

result of the dressing technique (the second pair of gloves is

worn after finalizing the dressing). Some aspects for aseptic

technique and behavior in the clean room are mentioned

in the FDA Guidance 2004. Very important for aseptic

manufacturing process are the detailed SOPs of the CDMO

such as aseptic operation, gowning room cleaning as well as

personnel and room environmental monitoring procedures.

Depending on the room classification or function,

personnel gowning may be as limited as lab coats and

hairnets, or as extensive as fully enveloped in multiple

layered bunny suits with self-contained breathing

apparatus. Cleanroom clothing is used to prevent

substances from being released off the wearer’s body

and contaminating the environment. The cleanroom

clothing itself must not release particles or fibers to prevent

contamination of the environment by personnel. Cleanroom

garments include boots, shoes, aprons, beard covers,

bouffant caps, coveralls, face masks, frocks/lab coats,

gowns, glove and finger cots, hairnets, hoods, sleeves

and shoe covers. The type of cleanroom garments used

should reflect the cleanroom and product specifications.

Personnel are the main

source of contamination

of clean rooms with

microorganisms. The

education and training of

the personnel, the

garments, the dress

procedures, the

rules for entry, and the

behavior inside the clean

rooms are important

factors (see EU GMP

Guide Annex 1, ISO 13408-

1, and FDA Guidance).

Establishing cGMP Manufacturing Capability for Phase 1 Sterile, Dispersed, Injectable Dosage FormsEstablishing cGMP Manufacturing Capability for Phase 1 Sterile, Dispersed, Injectable Dosage Forms

Page 17

Low-level cleanrooms may only require special shoes having

completely smooth soles that do not track in dust or dirt.

However, shoe bottoms must not create slipping hazards

since safety always takes precedence. A cleanroom suit

is usually required for entering a cleanroom. Class 10,000

cleanrooms may use simple smocks, head covers, and

booties. For Class 10 cleanrooms, careful gown wearing

procedures with a zipped cover all, boots, gloves and

complete respirator enclosure are required.

8. Process Simulation Validation

All manufacturing procedures in a pharmaceutical

manufacturing operation must be validated - according

to European Pharmacopoeia and FDA guidelines. This

is especially important for aseptic manufacturing of

parenteral dosage forms, where contamination poses a

significant patient risk. Process validation includes checks

on the process by means of process simulation tests using

microbial growth media (i.e., media fill tests). Since, in

pharmaceutical production, validated methods have been

already used for sterilizing equipment, processing air and

water and filtration techniques, media fill validation is very

much focused on the aseptic technique of the human

operator. Intensive training and education of personnel is

required in order to ensure that media fill validation is

recognized as a means of checking sterility level of aseptic

processing. According to all guidelines, process simulation

with media fill is state-of-the-art for validation of an

aseptic manufacturing process. Media fill means that a

microbiological nutrient media will be filled into a container

closure system (ampules, vials, etc.) instead of the

actual product. The filled container closure systems are

incubated under defined parameters and finally checked for

microbiological contamination. This is to demonstrate that

rooms, equipment and personnel are able to manufacture a

product with very low contamination rate.

Establishing cGMP Manufacturing Capability for Phase 1 Sterile, Dispersed, Injectable Dosage Forms

Page 18

The EU GMP Guide provides more details on this issue:

“Validation of aseptic processing should include a process

simulation test using a nutrient medium (media fill) ...

The process simulation test should imitate as closely as

possible the routine manufacturing process and include

all the critical subsequent manufacturing steps”.

The validation covers filling of media, environmental

monitoring, and incubation and evaluation of the filled

vials. Additionally the growth promotion properties of the

nutrient must be demonstrated. Microbiological examination

of positive vials, bio-burden examination of the materials

used and identification of contaminants are as well issues

that need to be considered. The simulation should consider

such conditions which simulate the highest risk (worst case)

of maximum expected and permitted loads. Examples for

worst case conditions are defined in ISO 13408. For example,

for vial dimension and filling speed, the worst condition is

the largest vial with the longest filling time. All interventions

and measures of the usual process should be simulated in

media fill. For example manual control of the filling volume,

change in personnel, and performance of environmental

monitoring. Even technical interruptions should be

considered (lack of air system, stopping of the filling

process, etc).

Liquid nutrient growth medium, capable of

supporting a wide range of microorganisms, is prepared,

sterilized, and filled in simulation of a normal manufacturing

process that includes compounding, sterile filtration,

in-process controls, sterilization of manufacturing process,

materials (garments, primary containers, filling equipment),

cleaning and sterilization process (e.g., cleaning in place -

CIP / sterilization in place - SIP) and filling. The sealed

containers of medium thus produced are then incubated

under prescribed conditions and subsequently examined

for evidence of microbial growth. If the media fill reflects

the standard procedure of product filling, the contamination

rate or contamination probability may be used as indicator

for the safety of the production process. Comprehensive

control of production environment, personnel, and

Establishing cGMP Manufacturing Capability for Phase 1 Sterile, Dispersed, Injectable Dosage FormsEstablishing cGMP Manufacturing Capability for Phase 1 Sterile, Dispersed, Injectable Dosage Forms

Page 19

installations, influencing the overall hygienic state of

manufacturing processes will be performed. Since, in

pharmaceutical production, validated methods have been

already used for sterilizing equipment, processing air and

water and filtration techniques, media fill validation is very

much focused on the aseptic technique of the human

operator. Intensive training and education of personnel is

required in order to ensure that media fill validation is

recognized as a means of checking sterility level of

aseptic processing.

Preparation of Media FillLiquid nutrient growth medium, capable of supporting a

wide range of microorganisms, is prepared, sterilized, and

then filled to simulate a normal manufacturing process that

includes compounding, sterile filtration, in-process controls,

sterilization of manufacturing process, materials (garments,

primary containers, filling equipment), and filling. The sealed

containers of medium thus produced are then incubated

under prescribed conditions and subsequently examined for

evidence of microbial growth. If the media fill reflects the

standard procedure of product filling, the contamination rate

or contamination probability may be used as indicator for

the safety of the production process.

Environmental monitoring, comprising airborne counts,

particle counts, and hygiene status of personnel and

materials - e.g., balances and compounding vessels -

is conducted during the weighing and compounding of

materials. Prior to filtration, the pH-value of the nutrient

broth is checked and in-process controls (IPC) on

identity, clarity, and bioburden are conducted. Samples

are controlled for analytical and microbiological controls

like that of any other product. Holding and process times

are documented and may be prolonged for validation

purposes. The bulk solution is sterile-filtered using the same

filter material as in normal aseptic processing. Filter integrity

is checked prior to and after use. Environmental monitoring

is conducted at this processing step. Prior to filling, primary

Media fills, or

process simulation

technique, is

generally accepted

as the procedure to

validate aseptic

manufacturing

processes.

Establishing cGMP Manufacturing Capability for Phase 1 Sterile, Dispersed, Injectable Dosage Forms

Page 20

containers are sterilized and depyrogenized, the filling line is

cleaned and sterilized (CIP/SIP) or transfer lines and dosage

pumps are sterilized separately.

Number of Fill Units and Filling Process DurationAccording to FDA Guidance the minimum number of filled

units of media fill is 3,000 units (to reach a confidence level

of 95% for demonstration of a contamination rate of less

than 0.1%). For batch sizes smaller than 3,000 units,

smaller numbers are acceptable (requirements are given

in ISO 13408 and EU GMP Guide Annex 1). For small batch

sizes (for example products used for clinical trials) at least

the actual batch size should be simulated during media fill.

For very large batches, it is recommended to simulate media

fill with 1% of the actual daily batch size. The vials with the

smallest and the biggest size should be regarded in media

fill. Table 2 gives the minimum units require required for a

media fill both for initial qualification and re-qualification

as well as the acceptable warning/action limits.

Table 2. First Qualification and Re-Qualification: Warning and

Action Limits in Media Fill According to ISO 13408-1

production batch number of units

First qualification

Requalification

minimum number of medium fill runs

minimum number of total filled units

warning limit / run

action limit / run

< 500

≥ 500 - 2999

≥ 3000

≥ 3

≥ 3

3

5,000

5,000

9,000

1

1

1

2

2

2

< 500

≥ 500 - 2999

≥ 3000

≥ 3

1

1

maximum batch size

maximum batch size

3,000

1

1

1

2

2

2

Establishing cGMP Manufacturing Capability for Phase 1 Sterile, Dispersed, Injectable Dosage FormsEstablishing cGMP Manufacturing Capability for Phase 1 Sterile, Dispersed, Injectable Dosage Forms

Page 21

Analytical Method ValidationThe extent of analytical procedures and methods validation

necessary will vary with the phase of the IND. Hence the

CDMO may elect to take a fundamental approach to

validation or qualification when appropriate. The main

goal of performing “staged’ validation in the early drug

development is to provide test procedures that are reliable,

able to support clinical studies, and evaluate the safety

of the product. The methods should use appropriate

parameters and sound scientific judgment, sufficient

information is defined as to ensure proper identification,

quality, purity, strength, and/or potency. The validation data

should be retained to link analytical procedures used in early

phase/pivotal clinical studies and a formal validation report

with change control may not be required. A brief summary

of the validation studies is recommended to be submitted

in the original IND.

The suitability of an analytical procedure (e.g., USP/NF, the

Official Methods of Analysis of AOAC International, or other

recognized standard references) should be verified under

actual conditions of use. Information to demonstrate that

USP/NF analytical procedures are suitable for the drug

product or drug substance should be included in the

submission and generated under a verification protocol.

The verification protocol should include, but is not limited

to: (1) compendial methodology to be verified with

predetermined acceptance criteria, and (2) details of the

methodology (e.g., suitability of reagent(s), equipment,

component(s), chromatographic conditions, column,

detector type(s), sensitivity of detector signal response,

system suitability, sample preparation and stability). The

procedure and extent of verification should dictate which

validation characteristic tests should be included in the

protocol (e.g., specificity, LOD, LOQ, precision, accuracy).

Considerations that may influence what characteristic tests

should be in the protocol may depend on situations such as

whether specification limits are set tighter than compendial

acceptance criteria.

Establishing cGMP Manufacturing Capability for Phase 1 Sterile, Dispersed, Injectable Dosage Forms

Page 22

Once an analytical procedure (including compendial

methods) is successfully validated (or verified) and

implemented, the procedure should be followed during

the life cycle of the product to continually assure that it

remains fit for its intended purpose. Trend analysis on

method performance should be performed at regular

intervals to evaluate the need to optimize the analytical

procedure or to revalidate all or a part of the analytical

procedure. If an analytical procedure can only meet the

established system suitability requirements with repeated

adjustments to the operating conditions stated in the

analytical procedure, the analytical procedure should be

reevaluated, revalidated, or amended, as appropriate.

Over the life cycle of a product, new information and

risk assessments (e.g., a better understanding of product

CQAs or awareness of a new impurity) may warrant the

development and validation of a new or alternative

analytical method. New technologies may allow for

greater understanding and/or confidence when ensuring

product quality. Applicants should periodically evaluate

the appropriateness of a product’s analytical methods

and consider new or alternative methods.

9. Conclusion

A small CDMO may choose to establish a GMP area

within a “laboratory setting” for the manufacture of drug

product in early development. Also, the use of isolators and

modular cleanrooms provides flexibility for adjusting the

manufacturing process to the product. Also, injectable,

sterile, disperable products can be manufactured aseptically

to avoid a terminal sterilization requirement. The FDA has

provided an amended rule that stresses that the cGMP

requirements of 21 CFR 211 are applicable only to Phase 2

and Phase 3 drugs and has provided guidance on the

requirements for small-scale Phase I cGMP manufacture.

Some of the regulatory burden is lifted for the manufacture

of Phase I materials as long as basic tenets of quality control

and validation are met.

Production of the

first clinical trial materials

for an injectable

pharmaceutical dosage

form is a significant

milestone, and a

challenging one for a

small company. CDMOs

focused on

manufacturing services

for Phase I sterile dosage

forms can provide a

reliable, cost-effective,

and timely solution to

this challenge.

Establishing cGMP Manufacturing Capability for Phase 1 Sterile, Dispersed, Injectable Dosage FormsEstablishing cGMP Manufacturing Capability for Phase 1 Sterile, Dispersed, Injectable Dosage Forms

Page 23

References

Center for Drug Evaluation and Research (CDER), Guidance for Industry, CGMP for Phase 1 Investigational Drugs, 2008.

European Directorate for the Quality of Medicines and Healthcare, European Pharmacopoeia (Ph. Eur), 8th Edition, 2016.

The Rules Governing Medicinal Products in the European Union, Volume 4, EU Guidelines to Good Manufacturing Practice, Medicinal Products for Human and Veterinary Use, Annex 1, Manufacture of Sterile Medicinal Products Brussels: 25 November 2008 (rev.) & 25 November 2010 (rev.).

United States Pharmacopoeia, Rockville, MD: <1211> Sterilization and Sterility Assurance of Compendial Articles, in USP 39, NF 34 (2016); <797> Compounding Sterile Preparations, in USP 39, NF 34 (2016); <1116> Microbiological Evaluation of Clean Rooms and Other Controlled environments, in USP 39, NF 34 (2016); <61> Microbial Limits in USP 39, NF 34 (2016); & <1208> Sterility Testing - V alidation of Isolator Systems in USP 39, NF 34 (2016)

CDER, Guidance for Industry Sterile Drug Products Produced by Aseptic Processing - Good Manufacturing Practice, 2004

International Standard Organization: Aseptic Processing of Health Care Products, Part 1, General Requirements, International Standard ISO 13408-1 (2015); & Cleanrooms and Associated Controlled Environments, Part 1, Classification of Air Cleanliness, International Standard ISO 14644-1 (2015)

Pharmaceutical Inspection Co-Operation Scheme (PIC/S), Recommendation on the Validation of Aseptic Processes, PI007-6 (2011)

Parenteral Drug Association (PDA): Process Simulation Testing for Aseptically Filled Products, Technical Report, No 22, (Rev 2011); Process Simulation Testing for Sterile Bulk Pharmaceutical Chemicals, Technical Report, No 28, (Rev 2006); Fundamentals of an Environmental Monitoring Program, Technical Report, No 13 ( Rev 2014); & Sterilizing Filtration of Liquids, Technical Report, No 26 (Rev 2008)

www.ascendiapharma.com

For more information contact:

Donald Pennino, [email protected]

Jim Huang, [email protected]

Biographies

Jingjun Huang, Ph.D., CEODr. Huang founded Ascendia in 2012 after fifteen years of

pharmaceutical R&D and management experience at Pfizer, Baxter,

AstraZeneca, and most recently Roche. Dr. Huang holds a Ph.D. in

Pharmaceutics from the University of the Sciences in Philadelphia.

He has lead the formulation development efforts for the successful

transition of several oral and parenteral dosage forms from

discovery through formulation, manufacturing, technical transfer

and ultimately commercialization. Dr. Huang’s research interests

are centered on improvement of solubility and dissolution, and

controlled delivery of, poorly water soluble drugs through

nano-emulsion and amorphous solid dispersion technologies.

He has been a reviewer for the Journal of Pharmaceutical Sciences,

International Journal of Pharmaceutics, Journal of Controlled

Release, Drug Development and Industrial Pharmacy, PDA

Journal of Pharmaceutical Science and Technology, Molecular

Pharmaceutics, and Pharmaceutical Research. Currently, he is a

member of American Association of Pharmaceutical Scientists

(AAPS) and American Chemical Society (ACS).

Donald Pennino, Ph.D., VP, QA/ARDDr. Pennino has over 45 years experience in the pharmaceutical,

biotechnology, and medical device industries. He holds a Ph.D. in

Physical Chemistry from Stevens Institute of Technology in Hoboken,

NJ. He has managed, designed and built many analytical, quality

control, and aseptic operation controlled areas over his career.

He has extensive validation expertise, as well as practical drug

development, helping to bring to market many products by

companies such as Novartis, Warner-Lambert, Schering-Plough,

Pfizer, Stryker BioTech, CR Bard, Chiron and Celator. He currently

is working on DoD projects to develop prophylactic vaccines as

defense to potential bioterrorism threats in the US. He also acts

as a consultant to industry providing regulatory solutions in

the area of quality, remediation and compliance.