AUTHOR OF THIS BLOG

DR ANTHONY MELVIN CRASTO, WORLDDRUGTRACKER

API SCALEUP (R AND D)

 
API Scale-Up During Research and Development
 
The ultimate goal of drug synthesis is to scale up from producing milligram quantities in a laboratory to producing kilogram to ton quantities in a plant, all while maintaining
high quality and reproducibility at the lowest cost. The term process in the pharmaceutical industry is broad and can apply to the process development work that leads to the efficient, reproducible, economical, safe, and environmentally friendly synthesis of the active pharmaceutical ingredient (API) in a regulated environment.
The increasingly stringent regulatory requirements and the global nature of the pharmaceutical business are continuously presenting new challenges to the pharmaceutical industry, resulting in increased competition and a need to produce high-quality APIs. API process development has subsequently gained more attention because of the potential to establish early control over the process at the research and development (R&D) stage by identifying and addressing problematic issues a priori. Thus, a systematic and prospective approach during R&D is key to achieving a successful prospective validation and scale-up. These activities are important and are frequently under scrutiny by the Food and Drug Administration.Prerequisites
The data generated in an R&D laboratory must be accurate, reproducible, and dependable. Therefore, it is imperative to establish and follow standard operating procedures (SOPs) for important activities such as the qualification and calibration of instruments and equipment (e.g., weighing balance, standard weights, temperature indicators, and reference standards). It also is necessary to keep proper detailed records of these qualification and calibration activities and other laboratory experiments, observations, and related analytical data.

 

 

Process considerations
API development. Current literature about the API and about its possible future developments should be kept in one place. Challenges to overcome at this stage include:

  • patent infringement;
  • inconsistent raw material quality and supply;
  • hazardous or nonregulated raw materials;
  • costly raw materials;
  • unsafe or environmentally hazardous reactions;
  • low yields;
  • difficult-to-achieve levels of purity (e.g., for enantiomers);
  • scale-up;
  • difficult-to-handle processes;
  • polymorphism-related issues;
  • stability of intermediates or products.

R&D chemists must devise a route that can address as many of these challenges as possible.

Cost. Raw materials, packaging materials, processes, and labor are major cost factors. R&D chemists can help reduce process expenses by:

  • suggesting cheaper alternative reagents or synthetic routes;
  • reducing raw material consumption (e.g., by conducting process-optimization studies);
  • shortening process time cycles;
  • recycling materials when possible.

Environmental friendliness. Today, R&D chemists are expected to use environmentally benign (i.e., green) chemistry. Ideally, high-yielding processes should be developed so that by-products are not pollutants or are treatable to eliminate pollution. Further processing of the spent materials should be attempted to recover the unreacted materials, by-products, and solvents. For example, a recovered solvent can be treated so that it can again match the desired quality specifications and thus be recycled in the same process step. Gaseous products should be scrubbed effectively. The final spent materials from the scrubber and the other processes should be assessed for their load on the environment and be handled appropriately, causing no environmental damage.

Solvent selection. The International Conference on Harmonization (ICH) guidelines have classified solvents on the basis of their risk to human health (1). Class 1 solvents should not be used during the manufacture of APIs. Such solvents include:

  • benzene (carcinogenic);
  • carbon tetrachloride (a toxic and an environmental hazard);
  • 1,2-dichloroethane (toxic);
  • 1,1-dichloroethane (toxic);
  • 1,1,1-trichloroethane (an environmental hazard).

Class 2 solvents should be limited because of their inherent toxicity. These compounds include:

  • toluene;
  • hexane;
  • methanol;
  • dichloromethane;
  • chloroform;
  • acetonitrile.

Solvents in Class 3 may be regarded as less toxic and of lower risk to human health. These include:

  • acetone;
  • ethanol;
  • ethyl acetate;
  • ethyl ether;
  • 1-butanol;
  • acetic acid.

Process adaptability. R&D chemists should modify their techniques to fit manufacturing environments. For example, to isolate a product, R&D chemists should avoid evaporating the solvents to dryness because it is difficult to follow such procedures in the plant. Instead, a suitable technique such as crystallization or precipitation should be developed because, in such cases, the product can be isolated by centrifugation or filtration in the plant. Similarly, the purification of a product should be achieved by means of crystallization or selective precipitation because other typical laboratory techniques such as column chromatography have operational limitations at the plant scale.

Methods of handling viscous materials in a plant also must be taken into account because the large surface area of plant equipment and piping can pose problems during material transfer. Solutions to these problems include performing one-pot reactions using a suitable solvent to transfer such materials. In addition, reactions involving low temperatures or high pressures could be difficult to handle in the plant, and an alternative route should be considered.

Safety precautions. Material safety. At the time of route finalization, R&D chemists should collect all raw material safety information—typically,material safety data sheets (MSDS). The storing and handling risks of these materials should be assessed, and appropriate measures should be taken to minimize them.

Process safety. During process development, significant consideration should be given to the safety of the chemistry being developed. The majority of industrially useful reactions are exothermic, thereby suggesting the need for risk assessment. Figure 1 shows the reaction enthalpies of two common reactions, indicating the high degree of process hazard associated with them .

The magnitude of overall heat release can be influenced by the kind of solvents used, concentration, other simultaneous processes taking place, and so on. Together, these factors may have enormous destructive power if they are not controlled properly. Therefore, all chemical processes that may be performed in a pilot plant should be subjected to a safety evaluation.

Initially, the thermal stability of the compounds (e.g., raw materials and intermediates) should be screened using differential scanning calorimetry to detect endothermic or exothermic behavior. Then, these results can be used to determine whether more careful and tedious measurements are required.

In some scale-up cases, an exothermic reaction can lead to thermal runaway, which begins when the heat produced by the reaction exceeds the heat removed. The rate of heat production may increase exponentially. Once control of a reaction is lost, the reaction vessel may be at risk of overpressurization caused by violent boiling or rapid gas generation.

Elevated temperatures may initiate secondary, more hazardous runaway or decomposition. Such a reaction hazard should be assessed in a laboratory using methods such as thermal gravimetric analysis (measuring the thermal stability of the reactants or products) and adiabatic calorimetry (measuring the decomposition
and release of gases). A process should not be performed in the pilot plant before
such safety assessment. Other process hazards such as dust explosion during milling
or storage may also be assessed at an early stage, depending upon the potential of
such risks.

Materials and vendors. It is important to conduct vendor audits and approval to ensure consistent quality and supply of raw materials, packaging materials, and other process components such as filtration media, gaskets, and O rings that might contact raw materials, process fluids, intermediates, or the API. Vendors should be selected according to criteria such as their market recognition, record, ability to supply consistent quality materials in time, and customer orientation.

Certificates of analysis (CoA) and MSDS of the materials should be obtained from vendors. Such information is useful while designing the specifications of raw materials and packaging materials and for obtaining recommended storage conditions.

Developing the specifications
In-house specifications can be developed on the basis of the results of user trials and the CoA of a vendor’s samples.

Process scale-up issues. It is important for R&D chemists to identify potential plant issues and to attempt to address these concerns suitably at the R&D stage. Laboratory studies such as those described below can help address many issues a priori to avoid surprises that might occur in the plant scale-up batches.

Simulating the R&D plant environment. Once the route is finalized, the plant environment in R&D should be simulated as far as possible by:

  • using reaction vessels of similar type and shape (e.g., material of construction, vessel shape, stirrer type, number of baffles, and diameter:length ratio of the vessel);
  • using the same charging sequence of the raw materials;
  • using similar mixing pattern and stirring parameters that are achievable in plant vessels (e.g., similar tip speed or power requirement per unit volume of the reaction mass that can be maintained in R&D);
  • developing suitable in-process sampling procedures that are feasible in the “controlled” environment of a good manufacturing practice plant;
  • using similar filtration cloth or medium;
  • using a similar type of dryer and drying parameters.

Such simulation experiments can help achieve better reproducibility at that plant scale because the possible deviations can be minimized.

Determining the scale-up factor. Many scale-up operations require more time than laboratory-scale experiments because the volume of materials is larger. R&D chemists should take into account the scale to which the process can be operated in the plant
and the required time cycles for such process steps. They should deliberately increase the process time cycles in a laboratory experiment to match the plant time cycles for similar operations. Process steps that may be considered for such studies include:

  • adding reactants;
  • mixing;
  • filtration;
  • centrifugation;
  • drying;
  • maintaining temperature.

The effect of such an increased cycle time on product quality and yield should be assessed. Thus, a scale-up factor at which the process can be operated without affecting the quality and yield can be determined prospectively.

Critical process parameters. While performing laboratory experiments, R&D chemists can test the limits of some operating conditions such as time, temperature, and pH or the quality parameter of key raw materials such as water content and impurity level. The effect of such challenges on product quality and/or yield should be assessed. If a parameter adversely affects either product quality or yield, it should be identified as a critical process parameter and be documented during the development stage.When scaling up, it is necessary to control such parameters strictly to ensure consistent product quality and yield.

Critical observations. During R&D experiments, it is important to record observations such as any signs of exothermic or endothermic activity during reaction, frothing, fuming, sublimation, pressure development, change in color, and change in phase. Similarly, observations related to reaction rate (e.g., vigorous or mild), filtration rate, flow characteristics of fluids, nature of the product (e.g., sticky, fluffy, amorphous, crystalline, semisolid) should be noted so that appropriate measures can be taken in the plant during scale-up.

Chemical compatibility studies. Certain process chemicals such as process fluids and intermediates may react chemically with manufacturing items such as process equipment, piping, flexible hoses, and filters while in direct contact with them, thereby leading to serious contamination and impurity issues.

R&D chemists should consider all process chemicals involved in API synthesis and obtain the data on their compatibility with various materials of construction of all processing items that may be involved during various operations  (3). In
the absence of such data, in-house data should be generated by simulating the exact contact conditions in a laboratory experiment, and the observations should be recorded.

For example, to determine the compatibility of a filter cloth with a process fluid, a sample piece of the filter cloth can be kept in contact with the process fluid for a specified time, temperature, or pressure in a laboratory, and the effect of such fluid on the weight, size, shape, color, elasticity, texture, and so forth of the sample cloth can be recorded. Such observations can help determine the suitability of using various processing items in the plant. During the purchase of such plant items, vendors
should supply material of construction certificates and chemical compatibility information.

Cleaning procedures. To develop prospective cleaning methods for plant equipment, suitable cleaning procedures for similar laboratory apparatus should be established. In a typical cleaning procedure, a similar flow pattern of cleaning solvents to that in plant equipment should be followed. Cleaning solvents such as acetone, methanol, and water should be selected on the basis of the solubility of the particular product that is to be washed from the empty apparatus. The visual observations regarding the effectiveness of cleaning and the analytical results of the rinse samples may facilitate the development of a prospective cleaning method for plant equipment during scale-up.

Stability data. Short-term stability data of the critical raw materials and intermediates should be generated in R&D under conditions similar to those in the plant. On the basis of these data, packaging and storage conditions for critical raw materials and intermediates can be established.

“Freezing” of specifications. As processes are fully developed and optimized, specifications of the in-process controls, intermediates, API, and the packaging materials can be “frozen.” That is, no change in the process should be allowed without change control assessment and approval.

Working standards. A working standard is a sample of highest purity that can be synthesized in R&D and purified to the maximum extent by repetitive crystallization or column chromatography. It then can be qualified by comparison with a suitable
reference standard such as a pharmacopeial reference standard. In the absence of such a reference standard, one should generate sufficient analytical data to support the structure of the working standard.

Stability. A representative sample of the R&D batch obtained by a “frozen” procedure should be kept for the stability studies. Therefore, early indications on the stability profile of future scale-up batches can be obtained, provided the same process is followed.

Other important challenges. In recent years, API process development has become more challenging because of the need for making APIs with a desired enantiomeric purity and a desired polymorphic form. The following examples justify the need for
the stringent regulatory requirements in these areas.

Enantioselective synthesis. More than half of the drugs used in clinical medicine are chiral compounds (4). Yet, the majority of these drugs are still prescribed as racemates. There is a need to make enantiomerically pure APIs when one enantiomer does not contribute to efficacy but may contribute to toxicity. Some interesting examples of isomers with various activities are shown in  (5).

The data  indicate a need for enantioselective synthesis in many racemic APIs. In such cases, one must validate the process carefully to ensure the formation of only the desired isomer. The other isomers should be considered impurities. In addition, it must be ensured that there is no chiral inversion during formulation, storage, or in the human body to avoid adverse side effects.

Synthesis of the desired polymorph. Polymorphism is the ability of a substance to exist in two or more crystalline phases that have different arrangements and/or conformations of molecules in a crystal lattice.Many APIs exist in several polymorphic forms with various properties such as crystallinity, bulk density, solubility, and bioavailability. Two interesting cases of product recalls that were a result of drug polymorphism follow (6).

Abbot Laboratories had to withdraw its HIV protease inhibitory drug Norvir (Ritonavir) from the market because an unwanted polymorph of the drug was produced (Form II) during shelf life. This form has a different dissolution rate to the known polymorph (Form I), thereby affecting the drug’s bioavailability.

Chloramphenicol-3-palmitate of Form B is a metastable form having eightfold higher bioavailability than the other polymorph, Form A, thereby creating a danger of fatal dosages when the unwanted polymorph is unwittingly administered as a result of alterations in process and storage conditions.

These examples demonstrate the need to identify all polymorphs of an API at the R&D stage. One can establish the polymorphs by determining physicochemical properties, by conducting thermodynamic stabilities, and by studying conditions of interconversions. Useful tools for such determinations include Fourier transform infrared spectroscopy, X-ray powder diffraction, and differential scanning calorimetry. The formation of a specific polymorph can depend on the type and composition of the solvents, temperature, synthetic route, storage conditions, and so forth. An interesting example of solvent composition giving various polymorphs is cholamide (7),which exhibits needle-like crystals (Form I) by recrystallization from a solution of 1:1 acetonitrile:water and platelet-like crystals (Form II) by recrystallization from a solution of 25:1 acetonitrile:water.

Once the desired polymorph has been identified, the process must be validated to obtain the desired polymorph consistently. Further, the stability protocol of the formulated drug must include some suitable tests to ensure that there is no change in the polymorphic form under these conditions.

Conclusion
The various process considerations described in this article can help chemists understand and adopt a systematic and prospective approach in research and development to have documented and controlled synthetic processes. This approach will help manufacturers meet product-quality objectives consistently and build a good basis for achieving the goals of prospective validation and scale-up activities.

References

  1. Impurities: Guidelines for Residual Solvents, Q3C, recommended by ICH on July 17, 1997.
  2. Process Chemistry in the Pharmaceutical Industry, K.G. Gadamasetti, Ed. (Marcell Dekker, Inc., New York, NY, 1999), p. 389.
  3. Internet databases such as Cole-Palmer Chemical compatibility database, ARO chemical compatibility, eFunda O ring material compatibility with chemicals, Varidisk chemical compatibility information, Flowline Chemical compatibility database and DMRTM fluid compatibility table by Daemar Inc.
  4. Physician’s Desk Reference (Thomson PDR,Montvale, NJ, 1997).
  5. S. Ahuja, Chiral Separations: Applications and Technology (ACS Publications,Washington, DC, 1996), p. 4.
  6. G. Chawla and A. Bansal, “Challenges in Polymorphism of Pharmaceuticals,” Scrip 5(1), 9 (Jan.–Mar. 2004).
  7. N. Yoswathananont et al., “A Novel Three-Component Pseudo-Polymorphism in the Cholamide Inclusion Crystals Promoted by the Combination of Organic Guest and Water,” Chem. Lett. 12, 1234 (2002).

 

 

Technology Transfer

Pharmaceutical manufacturers conduct more technology transfers now than ever before, yet technology transfer remains far from a core competency in the industry today. Looking at the technology-transfer practices of industry at 10 big pharmaceutical companies, the study found that most companies undertake more than 10 technology transfers per year—whether from development to commercial manufacturing or from one manufacturing site to another—and some execute many more. Many times industries does comprehend Process transfer as a Technology Transfer. Whereas there is a big difference which needs to be understand well before star up.

Process transfers vs. Technology transfers

Process Transfer is the transfer of process information, or capability, associated with process from a donor side (knowledge center) to a receptor side. The process is learned and realized by both sides and complies all the regulatory requirements in terms of Efficacy, Quality and Safety which is current Industry Practice. Whereas Technology Transfer, also called Transfer of Technology (TOT) is the process of skill transferring, knowledge, technologies, methods of manufacturing, samples of manufacturing to ensure that scientific and technological developments are accessible to a wider range of users who can then further develop and exploit the technology into new products, processes, applications, materials or services. It is closely related to knowledge transfer. In sight of recent QBD (Quality by Design) principles and approach the knowledge of Critical Quality Attributes (CQAs) of product which essentially be in line with Quality Target Product profile (QTPP) and effect of any change of Raw materials and critical process parameters (CMAs & CPPs) must be discussed & documented for future use. Any type of changes Deviations, Incidences, Change control and corrective and preventive action (CAPA) ought to realized and properly documented with the trends of CQAs & CPPs. Proper Risk assessment is mandatory exercise before any technology transfer activity.

……………

question In lab what are the factors needed to implement for active pharmaceutical intermediate

When developing active pharmaceutical intermediates in a laboratory, there are several critical factors to consider to ensure successful implementation of the process. Here are some of the most important factors:

  1. Raw materials: The quality of raw materials used in the laboratory must be consistent and of high quality. Any impurities in the raw materials can negatively affect the purity and quality of the final product.
  2. Process optimization: The process parameters, such as temperature, pressure, and reaction time, must be optimized to ensure that the product is produced with the desired purity, yield, and quality.
  3. Equipment: The laboratory should be equipped with the necessary equipment and instrumentation to carry out the process. The equipment must be calibrated and maintained to ensure accurate and reproducible results.
  4. Safety considerations: Safety procedures must be in place to protect laboratory personnel and ensure compliance with relevant safety regulations. Proper safety equipment, including personal protective equipment (PPE), must be provided.
  5. Quality control: Quality control measures must be in place to ensure that the product meets the required quality standards. This includes testing for purity, identity, and potency.
  6. Documentation: All aspects of the laboratory process must be documented, including raw material sourcing, process parameters, equipment calibration, and quality control measures. This documentation is important for ensuring reproducibility and traceability.
  7. Environmental considerations: Environmental impacts should be considered, and laboratory processes should be designed to minimize waste and reduce environmental impact.

Overall, implementing active pharmaceutical intermediate development in a laboratory requires careful planning, optimization, attention to detail, and adherence to safety and quality control measures.

questionDuring scaleup in pilot plant what are the factors needed to implement for active pharmaceutical intermediate

During the scale-up process for active pharmaceutical intermediates in a pilot plant, several factors must be considered to ensure the successful implementation of the process. Here are some of the critical factors:

  1. Equipment: The pilot plant should be equipped with the necessary equipment to carry out the scale-up process. The equipment should be capable of handling the increased volumes, and it should be compatible with the process requirements.
  2. Raw materials: The availability and quality of raw materials should be considered during the scale-up process. The pilot plant should be supplied with sufficient raw materials, and they should be of the same quality as those used in the laboratory.
  3. Process optimization: The process should be optimized for the larger scale to ensure that it produces the desired product with the same quality and purity as in the laboratory. The process parameters, such as temperature, pressure, and reaction time, should be optimized accordingly.
  4. Safety considerations: The pilot plant should be designed with safety in mind. Safety procedures and protocols should be put in place, and personnel should be trained on how to handle the process safely.
  5. Quality control: Quality control measures should be implemented to ensure that the product meets the required quality standards. This includes testing for purity, identity, and potency.
  6. Environmental considerations: The impact of the scale-up process on the environment should be considered. The pilot plant should be designed to minimize the environmental impact, and waste disposal procedures should be in place.
  7. Documentation: All aspects of the scale-up process should be documented to ensure reproducibility and traceability. This includes batch records, standard operating procedures, and analytical data.

Overall, the successful scale-up of active pharmaceutical intermediates in a pilot plant requires careful planning, optimization, and attention to detail to ensure that the final product meets the required quality standards.

 

question From Lab to Plant what are the factors needed to implement for active pharmaceutical intermediate

 

When transitioning from laboratory-scale development to plant-scale production of active pharmaceutical intermediates, several factors must be considered to ensure a successful implementation of the process. Here are some of the critical factors:

  1. Scale-up optimization: The process must be optimized for larger scale production, including scaling up equipment, optimizing process parameters, and addressing potential challenges, such as heat transfer, mixing, and mass transfer.
  2. Equipment: The plant should be equipped with the necessary equipment to carry out the scaled-up process. The equipment should be capable of handling the increased volumes, and it should be compatible with the process requirements.
  3. Raw materials: The availability and quality of raw materials should be considered during the scale-up process. The plant should be supplied with sufficient raw materials, and they should be of the same quality as those used in the laboratory.
  4. Safety considerations: The plant should be designed with safety in mind. Safety procedures and protocols should be put in place, and personnel should be trained on how to handle the process safely.
  5. Quality control: Quality control measures should be implemented to ensure that the product meets the required quality standards. This includes testing for purity, identity, and potency.
  6. Regulatory considerations: The plant should comply with relevant regulatory requirements, including good manufacturing practices (GMP), environmental regulations, and occupational health and safety regulations.
  7. Documentation: All aspects of the plant-scale process should be documented to ensure reproducibility and traceability. This includes batch records, standard operating procedures, and analytical data.

Overall, transitioning from laboratory to plant-scale production of active pharmaceutical intermediates requires careful planning, optimization, attention to detail, and adherence to safety, quality control, and regulatory requirements.

Take a tour   Indian food

North Indian Food vahrehvah.com

 

SAMOSA

.

 

THALI

 

/////////

40,414 total views, 1 views today

Share

 Leave a Reply

You may use these HTML tags and attributes: <a href="" title=""> <abbr title=""> <acronym title=""> <b> <blockquote cite=""> <cite> <code> <del datetime=""> <em> <i> <q cite=""> <s> <strike> <strong>

(required)

(required)

Time limit is exhausted. Please reload CAPTCHA.

Follow

Get every new post on this blog delivered to your Inbox.

Join other followers: