Search

Search
Generic filters

How to navigate the global chip shortage crisis if you buy pharmaceutical equipment

Thinking of purchasing new pharmaceutical equipment? These are our tips on how you can minimise the global chip shortage impact on your operations.

The global chip shortage crisis is already being felt across many industries. While most of the news has focussed on the auto industry, industries ranging from medical devices to home appliances and consumer electronics are experiencing a crunch. In this article, we provide a status update on chip shortages and pharmaceutical equipment manufacturers.

By Emma G McConnell, Science Reporter

What’s driving the global semi-conductor shortage?

In a nutshell, a surge in demand from the consumer electronics industry and supply-side disruptions.

With up to 75% of global chip supply coming from Asia, some of the supply-side disruption can be tracked back to COVID-19. But the problem of chip shortages in all equipment categories (automotive, medical, home appliances, etc) we’re experiencing runs deeper than the pandemic.

The main issue is one of raw material shortage. Imagine all the devices and equipment that use chips, not just the pc or laptop or mobile device you’re using to read this but countless other smart devices. Audio, Wi-Fi chips, imaging devices, process control modules, and displays. All these compete for the same semi-conductors and materials as they rely on the same technological underpinnings. So as chip usage has grown, raw materials shortages have become an increasing problem.

A few companies have exacerbated the crisis by stockpiling chips

It has been widely reported that a few companies—for example, in China—embarked on stockpiling chips in the hope of riding out the chip shortages. Unfortunately, this action has only compounded shortfalls, making it difficult for other equipment manufacturers to get in chips they require.

“Pharma equipment manufacturers are have reported no supply disruption”“Pharma equipment manufacturers are have reported no supply disruption”

 

What are the long-term consequences of chip shortages?

The good thing is manufacturers of pharmaceutical equipment have been aware of microchip shortages and have been exploring ways to avert any potential disruptions. All the companies we recently spoke to, including IMA, GEA and Agilent said they were not experiencing supply disruptions and delays. A sales manager at Freund Vector echoed the same sentiment when asked by PharmaCentral.

Things may be different with other companies so it’s important to assess how microchips are relevant to the company and how shortages may affect you as an equipment buyer. Speak to sales managers and ask them to confirm whether you may expect:

  • Longer wait times to have orders fulfilled and shipped
  • Specific brands shortages meaning that you may have to select alternative brand or model
  • Price increases

For now, the message we’re getting is that there’s no need to panic, even though many experts believe the impact may carry into 2022.

As a buyer of equipment, what practical steps can you take to mitigate the impact of pharmaceutical equipment microchip shortages?

PharmaCentral recommends several proactive steps you can take today to help minimize the potential impact of chip shortages and manage challenges.

  1. Plan well ahead

The number-one recommendation for buyers of pharmaceutical equipment is to plan ahead. If you expect needing equipment within the next 6 to 12 months, consider ordering it now—so you’re at the front of the line as companies work through their production backlog.

It is advisable to take time assessing whether you need to acquire new technology as part of your ongoing hardware lifecycle management, perhaps to support projected growth, or to have inventory on hand to replace non-functioning equipment. This prevents having to compromise or troubleshoot around a long-term delays .

 

  1. Factor in some delays

Even if you place an order now, fulfilment times may be longer than expected. So it’s advisable to plan for delays. Communicate with your vendor to have visibility of timelines, and manage expectations with your stakeholders accordingly.

 

  1. Be flexible

As a company or individual buyer, you may have a preference for specific brand or model of equipment. This is understandable. However, you should be open to being flexible if the brand or model of equipment you always buy is on backorder. Consider other brands and models even if they are not the exact model or brand you are familiar with so as to minimise disruptions.

 

  1. Evaluate other options

Purchasing hardware isn’t always the only option. Remember, the main reason for acquiring equipment is to support a specific process or objective. This business objective can be supported through leasing or outsourcing. The decision should be based on detailed assessment of requirements and all options carefully costed.

While pharmaceutical equipment chip shortages are yet to be felt, the global crisis will likely linger a little longer. It is likely that pharmaceutical equipment buyers will see some impact over the next several months. The message, though, is that if you’re looking to buy equipment, there is no need to panic. Be proactive, flexible, and think strategically ahead.

 

Formulating

European Patent Office 2020 Annual Review report published, reveals successful year, despite COVID-19 challenges

The European Patent Office published its Annual Review 2020 report on 29 June 2020. The report which was accompanied by a video, reveals a year of accelerated change as the organisation seeks to adapt to many challenges from COVID-19 pandemic.

The Annual Review 2020 shows that demand for European patents has remained nearly on a par with last year. A total of 180 250 European patent applications were received, representing a 0.7% drop compared with 2019. In addition, the Office published 133 715 European patents in 2020, -3% compared with 2019, but well above its target of 120 000.

The pandemic arrived at a time the Office was in the midst of implementing its Strategic Plan 2023. So it was necessary for the organisation to switch to a mostly virtual working model and be highly flexible. The review recognises the significance of these two factors to many of the main achievements and activities of 2020, which are outlined in the review under each of the Strategic Plan’s five clear goals.

You can find all details about this report on the EPO website or through this link.

 

Did you enjoy reading this story? Sign up to our free weekly newsletters and get stories like this sent straight to your inbox

 

How low-income countries can make generic Covid-19 vaccines

Vinod Kumar

PharmaCentral.com Guest Writers

Amid calls for developed nations to do more, US President Joe Biden recently announced that his administration will be availing 500 million doses of the BioNTech-Pfizer COVID-19 vaccine to 92 developing nations. The announcement was much welcome news, coming at a time when many low income countries are in a grip of rising coronavirus infections.

Low income countries are especially vulnerable to the pandemic due to, among other factors, their underdeveloped health systems and lack of local vaccine manufacturing capacity. If you consider that as of June 2021, of the 2 billion COVID-19 vaccine doses administered, only 0.3% has reached low-income countries, principally in Africa, where vaccination rates are as low as 1%. At this rate, it will take several years to achieve vaccine coverage that’s similar to that of wealthier countries.

So as much as Biden’s announcement is much welcomed, plenty of people are calling for a more sustainable solution. An official as COVAX, a UN scheme distributing jabs to poor countries, was recently quoted in the main press questioning whether the US pledge will have any impact at all. What is needed is expansion of vaccine manufacturing capabilities regionally, in Africa, Asia and Latin America, to produce generic vaccines quickly to avert needless deaths.

Problem is vaccine manufacturing is complex, and may not necessarily be cost-effective. It requires huge resources, expertise and time. In Africa, for instance, only Senegal is WHO prequalified to make vaccines. So for most of the continent, the option to repurpose existing facilities for local production does not exist.

The question on many peoples’ minds then is, what option do low income countries have? Is local vaccine manufacturing viable? Admittedly, this is vast topic and my is to provide an overview rather than a deep dive analysis.

 

What it takes to manufacture vaccines

In short, a lot! First, here are the headline figures: estimates very but generally between US$ 200 M to 700 M to develop a vaccine from concept to market, and another USUS$D 50 M – 130 M to construct, equip and commission the manufacturing facility. Development time and facility construction take at least 3 years, plus a further 1 year lead time to get the vaccine to market after commissioning.

Conceptual vaccine development process (adapted from multiple sources, including Robinson, 2016)

 

Money aside and perhaps most important, many things must perfectly align. Robust and well defined methods and processes have to be developed, testing and monitoring regimes established, and above all, a company-wide commitment to comply with internal and external standards and regulations established. It is why vaccinologists say “the vaccine is the process” referring to the sum total of different steps that define success or failure.

So while not impossible (COVID-19 has taught us what science can achieve), it is no small feat, particularly for a country with no prior manufacturing experience.

 

Basics of vaccine manufacture

There is currently no standard vaccine manufacturing process. Processes differ by vaccine platform and at times, from manufacturer to manufacturer. However, for most vaccines, the process involves some sort of cell culture/fermentation, isolation and purification of active substances, formulation, fill-and-finish, and packaging. The entire process, from start to finish can take anywhere between 7 days to several months.

If we take a biological-based vaccine, we can categorise the different manufacturing processes into three main steps: in the first step (upstream), the cell culture is developed, standardised and induced to produce the active substance (e.g protein or virus). In the second step (downstream), the cell culture is harvested, purified and filtered to produce the pure active substance. In the last step (fill-and-finish), the active is formulated (excipients and stabilisers added) into a vaccine, filled into vials, inspected and packaged ready to ship to the clinic.

The process for the manufacture of mRNA vaccines is less complex because it is mainly synthetic. It starts with a plasmid DNA for the mRNA being produced via fermentation. Using an enzyme-catalysed in-vitro transcription process, the mRNA that codes for the spike protein is produced. The new mRNA is chemically modified (capping and stabilisation) to render it biologically active. It is purified and then formulated into lipid nanoparticles in a microfluidic process. Once formulated, it undergoes fill and finish, quality control and packaging in the same way as other vaccines.

 

What potential obstacles would a new manufacturer face?

Vaccines are not created equal, but you knew that already. The range and relative production complexity of several vaccine types is shown below. At one end is the live attenuated oral polio vaccine with significantly lower complexity and production costs while at the other end is the pneumococcal conjugate vaccine.

Vaccine types and comparative production complexity

 

Notwithstanding, there are common equipment or equipment types shared across platforms such as bio reactors, filtration and purification, filling and lyophilisation equipment, although the sequence of operations and the specific cycles for each product may vary. In most cases, each product has its own dedicated facility requirements as well as skillset.

Here below are some of the most important considerations:

1. Process development and maintenance

Introducing new or changes to facilities, manufacturing equipment, processes or raw materials triggers regulatory examination to confirm that processes are robust enough to ensure the vaccine is equivalent to the product produced in the original clinical or reference batches. Erecting a new facility and the accompanying processes will be a significant undertaking because the facility and processes define the product, but also all stakeholders have to have visibility into how the manufacturing process, quality control, specifications as well as all the support utilities will pan out at commercial scale quite early one.

 

2. Raw materials and consumables

Many of the raw materials and consumables used in vaccine manufacture are highly specialized in nature, with a few being produced by biological production methods. For these reasons, there are only so many suppliers available and the supply situation is subject to shortages or long lead times, not helped that we are currently in the middle of a pandemic and pressure on supplies is heightened. When materials are in short supply they tend to be expensive due to usual supply and demand dynamics. One option is to qualify multiple suppliers especially for critical materials, but this means extra work auditing suppliers and qualifying materials, which likely increases costs of goods.

 

3. Regulatory affairs and commercialization

Vaccine regulatory requirements and steps for obtaining marketing approval are well documented. They are also broadly similar across the world although the exact compliance requirements differ from country to country. Also, some vaccine products may be made only for specific countries based on their requirements, and for these, regulatory agencies may have their own flexibilities built into review and inspection regimes.

Whatever the local arrangements, manufacturers are expected to comply with all requirements applicable for the relevant regulatory agencies (including those the company wishes to sell its vaccine). These requirements include adherence to good manufacturing practices as well as routine monitoring of adverse event data, and annual reporting of specific manufacturing information (e.g., data trends, change management, stability review, CAPA, etc).

In addition, a manufacturing facility will be routinely subjected to external audits and inspections to assess compliance with GMP, facility maintenance, manufacturing and quality systems, and performance of the process. For the uninitiated, this is a lot to take.

If the new company wishes to sell their vaccine to highly lucrative WHO and UNICEF projects (essential for long-term business sustainability), then compliance with WHO’s Pre-Qualification (PQ) is mandatory. A PQ assessment process can take up to a year to complete excluding any additional required to respond to queries.

But even before a vaccine can be considered for PQ, the sponsoring regulatory agency must be first qualified as “functional” by WHO. WHO’s standards for “functionality” are immense and currently, there are only two African states with an national regulatory agency adjudged as “functional” for vaccines sponsoring purposes.

 

Manufacturing costs

The costs of manufacturing a vaccine are influenced by several factors. Here, I will focus on facilities, equipment, raw materials, process development and labour but clinical development and financing costs. Facilities and maintenance, raw materials, production, personnel and compliance comprise direct costs, while indirect costs include the creation and implementation of quality systems, operations planning, inventory and distribution, and sales, marketing and management. In the table below, we list some of the important costs and how they could be reduced.

 

Major vaccine production cost drivers and ways to reduce them

 

Major Cost Driver Impact on overall costs Cost range Examples of ways production costs can be lowered
Product Development

R & D facilities

R & D staff

Research costs

High

(High fixed costs)

>500 M US$ Copy originator process where possible

Perform technology transfer

Use immunological surrogates in lieu of efficacy studies

Purchase antigens and execute form/fill prior to full end-to-end manufacturing

Facilities and Equipment

Land

Buildings

Equipment

 

Ongoing maintenance costs

High

(high fixed costs)

50 to 130M US$ Design for very high facility utilization

Repurpose existing facilities

Use multi-dose vials

Use single-use disposable systems

Consider blow-fill-seal technology

Use closed systems to reduce classified production space

Direct Labour

Wages & benefits

Low Comparatively lower in developing counties (typically 25 – 50%) Increase single-use production technologies

Develop capacity progressively

Overhead

Management, quality systems, IT systems

High Up to 50% cost of raw materials and labour Streamlined quality systems.

Management with broad expertise

Licensing/Regulatory and commercialization High 100k to 1M US$

For staff and consulting costs, WHO PQ, Site audits & evaluation fees

Pursue WHO PQ only if needed

Request royalty reductions or waivers

Accelerate approval by seeking NRA or WHO priority review for emergency use

 

Facilities and equipment maintenance

Constructing and maintaining a vaccine manufacturing facility is a major cost for a vaccine manufacturer. A green-field, purpose-built vaccine manufacturing facility in North America can easily cost 50 – 750 M US$ per antigen, depending on the complexity of design, automation, segregation, utilities, and contamination controls. Based on current rates, GMP space cost from 6k US$/m2; while non-GMP space costs 3k US$/m2. The cost of clean rooms and containment rooms is higher. It can take up to 7 years to design, build, validate, and commission a manufacturing facility. These estimates are for developed countries, so actual facilities cost may be lower in developing countries, although not by much since many materials and fixtures need to be imported and some key personnel hired from other countries as expatriates.

 

Process development

The focus here is on the steps used to develop and validate the manufacturing process leading to a licensed product. The key steps for process and analytical development and associated time frames during clinical development are outlined in the table below.

 

Key vaccine development stages and process/system expectations

 

Phase Goal Remark
Exploratory & Pre-clinical Immunogenicity & safety assessment of target antigen or cell in cell culture or animal disease model Small scale. Attention paid to method of manufacturing as it impacts nature of vaccine. Process development not critical at this stage
Clinical Trial Authorization Application Obtain approval to conduct human clinical studies An outline critical manufacturing and analytical methods for producing vaccine required. Also, quality, purity and stability required to be demonstrated
Phase I Vaccine Trials Safety assessment of candidate vaccine. Type and extent of immune response elicited GMP manufacture of all clinical trial material. Process development ideally optimised by this stage although this can be deferred
Phase 2 Vaccine Trials Assessment of safety, immunogenicity, dose response, schedule of immunizations All major process changes completed and qualified. Anticipated cost of goods determined
Phase 3 Vaccine Trials Assessment of candidate in target population for safety & adverse events. Efficacy is estimated. manufacturing consistency confirmed Processes finalized and validated. Analytical tests for release are completed and validated. Costs of goods are confirmed as any changes need to be re-validated and may require additional clinical testing
Approval & Licensure Biological Product Application approval Completed full review and details of manufacturing methods and analytical methods; shelf-life stability studies; process validation, facility validation, release testing validation; launch lots prepared and released. Regulatory agency inspection of all manufacturing and release facilities and manufacturing and quality systems documentation

 

Costs of process and analytical development, manufacturing, and documentation are highly dependent on vaccine type, technical complexity, disease target and regulatory body. Some independent analysists have calculated that the Chemistry, Manufacturing and Controls (CMC) development aspects for a vaccine range from 5 M US$ to 50 M US$ and require between 50 and 100 person-years in human resources. If a manufacturer from a developing country chooses to licence technology or partner with an established manufacturer, this can reduce costs and development time significantly.

 

Labour costs

Having a motivated, technically competent and committed workforce is a major requirement for any vaccine manufacturer, irrespective of where there are domiciled. Companies also need to be able to hire, train, and develop their workforce, a challenge even for highly experienced manufacturers.

For many reasons we don’t have time and space to go into, there is a shortage of personnel with the requisite skills and expertise needed by the vaccine industry. The deep scientific knowledge required to support such an endeavour takes years to acquire, so developing countries are at a disadvantage when it comes to establishing and sustaining local vaccine manufacturing capacity.

Countries such as China and India have large populations to draw from, not mentioning the huge financial resources, which have enabled them to build relatively sound technical and scientific foundations within a single generation. It is not an accident that these countries have also successfully managed to enter into vaccine manufacturing. Other developing countries are not as well endowed, so developing a knowledge base in tandem with a comprehensive training system within a short period of time is not as easy.

Finally, staffing for an average facility in low-income country will often include local and several expatriate employees in order to entrench and safeguard the relevant technical skills. Expatriate staff, by their nature, require higher total compensation, which increases overall labour costs.

 

What options are available to developing countries?

The good thing is that the pace of technology and innovation now mean that production costs need not be a showstopper even for smaller, resource-poor countries. With small-scale, modular or disposable technologies, high-density bioreactors, and innovations in fill-and-finish processing it is possible for new entrants to successfully venture into vaccine manufacturing.

For low income countries, the proven route is starting with fill and finish capacity, and then through a phased approach, step up the value chain to manufacture antigens as well. This approach allows reduction of upfront investment risk while building manufacturing know-how in a controllable way.

As we mentioned earlier, not all vaccines are created equal. This also applies to building vaccine capacity. Biological-based production technologies (such as those for recombinant-protein or viral-vector vaccines) have higher capital and operational costs compared with novel mRNA-based vaccines, which can be easily and quickly synthesized in a chemical reactor. Further, mRNA-based processes will soon become even more accessible through mobile “RNA printers” that are promised to further reduce footprint, labour and cost commitments.

To negate the high up-front costs of purpose-built facilities, we recommend using modular and prefabricated facilities, which constructed off-site and delivered to the site where they will be put into use manufacturing. Prefabs have been used for many years in small molecule pharmaceuticals and offer benefits of fast delivery and capacity flexibility. They are now available for vaccine manufacturing, with delivery times of three to six months instead of 5 – 7 years.

An example of prefab solution is the one Exyte and Univercells Technologies have co-developed and is available for vaccines. Known as NevoLine, this system uses continuous/semi-continuous process equipment with automation, and a small manufacturing footprint which can be housed inside the ExyCell cleanroom. The facilities can be commissioned fairly quickly, typically within 3 months.

Finally, it is no secret that labour productivity is a major limitation in low income countries, but more so in many African countries. For these countries, productivity can be boosted through pool human resources and technology transfer from across the value chain – including from equipment and raw material vendors, to universities and NGOs. For instance, organisations such as WHO and PATH have platform which provide working pilot plant production processes (including all the SOPs, documentation and training), allowing new entrants to establish a robust production system fairly quickly while managing to reduce development costs and improve productivity.

So there we have it. Vaccine manufacturing is a capital-intensive endeavour. Short and long-run product costs are primarily driven by development and production-related costs, with facility maintenance, compliance with regulations, raw materials and labour being the most significant contributors. Countries seeking to localize vaccine supply need to invest heavily in facilities, equipment, skilled labour and ongoing quality management with a long time-horizon.

But investment in facilities, equipment and skilled labour is not enough. Having sound scientific knowhow is critical to vaccine production, so the issue of methodical transfer of knowhow, the possession of which provides the basis for long-term viability of the venture, becomes especially important. In this sense, technology transfer is one of the main ways ventures in low income countries can possibly acquire vaccine manufacturing capacity.

 

References

  1. Milstien J., Batson A., Meaney W. A systematic method for evaluating the potential viability of local vaccine producers. 1997;15:1358–1363. [PubMed] [Google Scholar]
  2. Robinson J.M. Vaccine production: main steps and considerations. In: Bloom B., Lambert P.H., editors. The vaccine book.2nd ed. Academic Press; San Diego: 2016. pp. 77–96. [Google Scholar]
  3. Gerson DF, Mukherjee B. Manufacturing process development for high-volume, low-cost vaccines. BioProcess International. April 2005.
  4. Gomez PL, Robinson JM, Rogalewicz JA. Vaccine Manufacturing. Vaccines, 6th ed. In: Plotkin S, Orenstien W, Offit P. Orlando, editors. WB Saunders Company; 2013. p. 44–57.
  5. Wilson P, Giving developing countries the best shot: an overview of vaccine access and R&D; 2010.
  6. WHO. WHO Prequalification Financing Model – Questions and Answers <http://www.who.int/medicines/news/prequal_finance_model_q-a/en/> [accessed 17.05.05].
  7. WHO. WHO updates on regulatory system strengthening and prequalification activities; 2016. <https:///www.unicef.org/supply/files/RSS_and_PQ__updates_vaccines-ID_c_rodriguez.pdf> [accessed 17.02.07].
  8. Sinclair A, Latham P. Vaccine production economics. In: Wen EP, Ellis R, Pujar NS, editors. Vaccine Manufacturing and Development; 2015. p. 415.
  9. Plotkin S, Robinson JM, Cunningham G, Iqbal R, Larsen S. The complexity and cost of vaccine manufacturing – An overview. Vaccine. 2017;35(33):4064-4071. doi:10.1016/j.vaccine.2017.06.003

 

Your FAQs Answered: What you need to know about Aducanumab, the recently approved Alzheimer’s drug from Biogen

Last Monday, the US Food and Drug Administration granted approval for Aducanumab, the first new drug treatment for Alzheimer’s disease in 2 decades. Here, I attempt to provide answers to common questions on this landmark new drug, also known as Aduhelm.

What is Aducanumab?

Aducanumab is a monoclonal antibody therapeutic. Monoclonal antibodies are man-made proteins designed to act like human antibodies. Aducanumab is designed to bind to and help destroy amyloid beta (proteins) from the brain. Amyloid beta is thought to be involved in the progression of Alzheimer’s disease.

How does it work?

Aducanumab works by eliminating beta-amyloid plaques in the brain — a process that, according to Biogen, slows down the ability to destroy brain cells. Whether this alone is enough to slow down cognitive decline is not clear for now. Assessments, based on tests of cognition and function, for Aducanumab and a placebo showed only marginal improvement.

Who can get the drug?

Aducanumab was approved using the accelerated approval pathway. The drug was tested in patients with mild cognitive impairment, so is now approved for all patients with Alzheimer’s. But there is a catch. Post-approval trials will still be needed. Patients will need regular monitoring after starting treatment) and if post-approval trials fail to show the drug works as intended it could be pulled from the market.

How much does it cost?

According to Biogen the yearly cost for a maintenance dose, based on an average patient’s weight, will be $56,000. This is the list price, not the price paid by patients or government agencies like the NHS. The final cost of treatment will depend on many other factors.

How is the drug administered?

Aducanumab is given by IV over about an hour once every four weeks.

What can patients expect?

Aducanumab is not a cure and it does not reverse the disease’s progression. In clinical trials, success was measured not by cognitive improvement but by slowing in the rate of cognitive and functional decline.

Any side effects?

In two clinical trials, about 40% of clinical trial patients who got the approved dose of Aducanumab developed painful brain swelling. Symptoms included headache, dizziness, visual disturbances, nausea, and vomiting; about 17% to 18% of patients had microhemorrhages in their brains.

When will the drug be available?

Biogen expects to start shipping supplies in about two weeks across the U.S. International shipments will likely take longer pending approval, if and when it happens.

Why has this drug been so controversial?

For several reasons: First, Biogen had to stop two trials of Aducanumab in March 2019 after independent monitors, looking at data during an interim analysis, concluded the drug was unlikely to benefit patients. Second, and perhaps most importantly, the amyloid hypothesis, on which this drug is premised, has yet to be proven. It’s not clear if amyloid plaques, or tangles of another protein called tau, are causes or effects of Alzheimer’s. Finally, many experts are concerned that the desperate need for new treatments might be leading regulators to accept limited evidence of efficacy.

What other drugs are available now?

Three acetylcholinesterase inhibitors, donepezil (Aricept), galantamine and rivastigmine and memantine (Ebixa), a glutamate receptor antagonist, are all approved for managing mild to moderate Alzheimer’s disease.

What else is in the pipeline?

As of May 2021, there are over 120 agents in clinical trials. Most drugs in trials aim to achieve disease modification by targeting the underlying biological processes of Alzheimer’s disease (AD).

The Future of Science Events Post-COVID?

Excipients and Formulation Approaches Employed in Leading Covid-19 Vaccines

In this article, I compare excipients and formulation methods used in the four Covid-19 vaccines from Pfizer BioNTech, Moderna, Astra Zeneca and Janssen-Cilag (Johnson & Johnson), that have obtained emergency approval by the UK’s Medicines and Healthcare products Regulatory Agency (MHRA) and its European counterpart, the European Medicines Agency, EMA.

Introduction

SARS Covid-19 has put vaccines into the public limelight again. Now more than before, we are all aware of the need for and challenges of timely distribution, as well as the importance of the cold chain. What is perhaps not clear to most people is that these challenges are heavily influenced by a vaccine’s inactive ingredients (excipients, solvents & adjuvants) as well as the platform.

In addition to stability, a vaccine’s formulation and excipients impact the economics of manufacture and the finished product’s presentation. It is the same way an engineer at Ferrari or Aston Martin designs an impressive V8 powertrain but finds that he or she still needs an equally impressive chassis for the new engine to deliver the performance.

This is why an understanding of how vaccines are formulated and the reasons behind choice of different excipients, from a pharmaceutical technology perspective, is equally important to appreciating differences in manufacturing, storage and distribution requirements.

First, I will briefly provide an outline of the vaccine platforms currently in use as this is important to formulation and excipients selection.

Vaccine Platforms

There are any number of ways to categorise vaccines. One method I like to use is classify them by the technology or platform used. Using this approach, we can distinguish four main platforms:

Whole pathogen vaccines

This is the oldest and most well-known method of vaccine development. It involves using an entire disease-causing organism in the vaccine to elicit the immune response, analogous to that obtained in regular infection. Whole pathogen vaccines are further divided into live attenuated and inactivated vaccines.

In live attenuated vaccines the disease-causing organism is weakened (attenuated) to curtail its disease-causing ability although it’s still able to replicate and trigger an immune response. An example is the Oral Polio Vaccine.

Inactivated vaccines have the genetic material destroyed – this way they are not able to replicate and infect cells, but are still able to trigger an immune response. Since inactivated vaccines do not always create a strong immune response as live attenuated vaccines, adjuvants (for example, aluminium hydroxide and aluminium phosphate are included in the formulation. An example is the Hepatitis A vaccine.

Subunit vaccines

These vaccines typically contain one or more immunogens from the surface of the pathogen. Antigens are usually produced through recombinant technologies. Subunit vaccines can be further divided into recombinant protein vaccines; toxoid vaccines, conjugate vaccines, virus-like particles and outer membrane vaccines.

The vast majority of vaccines in use today are subunit vaccines – they do not contain any whole bacteria or viruses and instead contain polysaccharides or proteins or their combination from the surface of bacteria or viruses, which are recognised by the immune system.

Agencies such as the World Health Organisation and the CDC, attest to the excellent safety profiles of subunit vaccines. Their only ‘downside’ is that they often require inclusion of adjuvants. Examples of subunit vaccines include

Nucleic acid vaccines

Nucleic acid vaccines work by providing genetic codes for host cells to produce antigens, which then stimulate the immune response. Nucleic acid vaccines can be further divided into RNA and DNA vaccines.

RNA vaccines use mRNA which is formulated in a lipid nanoparticle for protection and fusion with the cell membrane. A drawback of RNA vaccines is their inherent instability.

DNA, being more stable than mRNA, doesn’t require the same initial protection. DNA vaccines are typically administered using electroporation to allow cells to take up the DNA. There are currently no licenced DNA vaccines, however there are several in different stages of development.

Viral vectored vaccines

Viral vectored vaccines utilise harmless viruses to deliver the genetic code of target vaccine antigens to cells of the body, so that they can produce protein antigens to stimulate an immune response. Viral vectored vaccines can be developed quickly and on a large scale. They are also significantly cheaper to produce compared to nucleic acid or subunit vaccines.

Viral vectored vaccines can be further classified into replicating and non-replicating. In the former, viral vectors retain the ability to make new viral particles alongside delivering the vaccine antigen when used as a vaccine delivery platform. Non-replicating, as the name suggests, do not retain the ability to make new viral particles because some of the viral genes required for viral replication have been removed.

Differences by Vaccine Platform

The Pfizer BioNtech and Moderna vaccines are nucleic acid vaccines. Both the Pfizer BioNTech Covid-19 vaccine (BNT162b COVID-19 mRNA vaccine) and the Moderna Covid-19 vaccine (mRNA-12743 COVID-19 vaccine) are single stranded, 5’ capped messenger RNA produced by cell-free in vitro transcription from corresponding DNA templates that encode for SARS-Cov-2 spike protein.

Although mRNA vaccines are a relatively new technology (approx. 30 years old, compared to whole organism vaccines that were first introduced during the late 1700s), they are well studied. They also offer many advantages:

  1. Firstly, no live components are involved, so there is no risk of the vaccine triggering disease.
  2. The mRNA, due to its transient nature, also presents zero risk of becoming integrated with our own genetic material.
  3. Moreover, the immune response involves both B and T cells.
  4. Finally, and perhaps more importantly, they are relatively easy to manufacture.

The major downsides of mRNA vaccines are that they often require ultra-cold storage, and almost always require booster shots for maximum effectiveness. We will touch on this later in the article.

By comparison, the Jansen-Cilag and AstraZeneca vaccines are viral vectored vaccines, so they use a different approach to instruct human cells to make the SARS-2 spike protein. The Jansen-Cilag Covid-19 vaccine (COVID-19 vaccine (Ad26,COV2-S [recombinant])) uses a non-replicating adenovirus (a Novel Adenovirus Type 26) from their AdVac technology and grown in PER.C6 cell line.

The AstraZeneca Covid-19 vaccine (COVID-19 (ChAdOx1-S [recombinant]) also uses a non-replicating adenovirus, this one being a chimpanzee adenovirus (as opposed to Ad26, which is human adenovirus) known as Oxford1 (or ChAdOx1).

A schematic illustration of an adenovirus vector vaccine is shown below:

Fig. 1: Schematic illustration of an adenovirus vector vaccine

Fig. 1: Schematic illustration of an adenovirus vector vaccine

The adenoviruses used in these vaccines are engineered to only carry the genetic code for the SARS-2 spike protein. Upon entering human cells, they use that code to make spike proteins. These vaccines mimic natural infections, which is advantageous in triggering strong cellular immune responses as well the production of antibodies by B cells.

The technology is well-established, with two other vaccines already approved (Ebola & Zika vaccines). However, adenovirus vaccines are relatively complex to manufacture, and with time, their effectiveness reduces.

How Covid-19 Vaccines Differ in Formulation and Excipients Used

Pfizer BioNtech and Moderna Vaccines

The Pfizer BioNtech and Moderna vaccines are available as sterile, multi-dose colloidal dispersions for intramuscular injection. The mRNA in both vaccines is encapsulated in lipid nanoparticles (LNPs). LNPs are chosen to overcome the inherent hydrolytic instability, poor membrane permeability, and the abundance of RNAses in the body.

Lipid nanoparticles (sometimes called solid lipid nanoparticles, SLNs) are colloidal carriers made from lipids. As a drug delivery technology, LNPs emerged in the early 1990s as an alternative to traditional emulsions and liposomes. Although their exact structure is still under debate, LNPs are generally thought to consist of a solid lipid core (unlike liposomes which have an aqueous core) and an external phospholipid layer (membrane). A schematic illustration of an LNP is shown in figure 2. You can also watch an excellent YouTube on LNPs through this link.

Fig. 2: Schematic illustration of an lipid nanoparticle vaccineFig. 2: Schematic illustration of an lipid nanoparticle vaccine

For the Pfizer BioNtech and Moderna vaccines, LNPs are obtained by admixing mRNA, various lipids, which include a neutral phospholipid, cholesterol, a polyethylene-glycol (PEG)-lipid, and an ionizable cationic lipid (which has amine groups (at low pH) and facilitates interaction with the anionic mRNA during particle formation and also membrane fusion during internalization). The PEG-lipid controls particle size and acts as a steric barrier, preventing aggregation during storage. When complexed with the mRNA, the LNPs-mRNA particles have sizes in the range of 60–100 nm.

Table 1 below summarises the main differences in Pfizer BioNtech and Moderna vaccines’ formulation and excipients:

Pfizer-BioNTech vaccine Moderna vaccine
Name of product Comirnaty mRNA-1273
Active BNT162b2 (single-stranded, 5’ capped mRNA) mRNA 1273 (single-stranded, 5’ capped mRNA)
mRNA dose; route of administration 30 µg; intramuscular 100 µg; intramuscular
Delivery system Lipid nanoparticle made from

ALC-0315 or 4-hydroxybutyl) azanediyl)bis (hexane-6,1-diyl)bis(2-hexyldecanoate), ALC-0159 or 2-[(polyethylene glycol)-2000]-N,N ditetradecylacetamide; 1,2-Distearoyl-sn-glycero-3-phosphocholine (DSPC) and Cholesterol

Lipid nanoparticles made from

SM-102 (heptadecan-9-yl 8-((2-hydroxyethyl) (6-oxo-6-(undecyloxy) hexyl) amino) octanoate}; PEG2000-DMG = 1-monomethoxypolyethyleneglycol-2,3-dimyristylglycerol with polyethylene glycol of average molecular weight 2000; 1,2-Distearoyl-sn-glycero-3 phosphocholine (DSPC) and Cholesterol

Diluent Water for injection

Buffered by Potassium dihydrogen phosphate, Disodium hydrogen phosphate dihydrate

pH 7–8

Water for injection

Buffered by Tris (tromethamine)

pH 7–8

Other excipients Potassium chloride

Sodium chloride

Sucrose

Sodium acetate

Sucrose

Table 1: Formulation of Pfizer BioNtech and Moderna COVID-19 vaccines

LNPs are particularly unstable thermodynamically. In addition, they are susceptible to chemical instability, which can arise from hydrolysis and oxidation of the lipids in the LNPs, as well as oxidation of unsaturated fatty acid groups. This makes LNPs systems especially susceptible to storage conditions, which helps explain, in part, to the stringent handling conditions required of mRNA vaccines.

Janssen-Cilag (Ad26. COV2.S) and AstraZeneca (Vaxzevria or AZD1222)

Janssen-Cilag (Ad26. COV2.S) and AstraZeneca (Vaxzevria) vaccines are available as sterile, multi-dose aqueous suspensions for intramuscular administration. Liquid suspensions are an efficient and the go-to format for viral vector gene delivery systems. However, the challenge faced by formulators is ensuring long term stability since, unlike conventional pharmaceutical products, they are complex biological structures susceptible to chemical and physical stressors, such as changes in solution pH, ionic strength, redox potential and surface activity.

Thus, the aim of formulation efforts here is to prevent conditions likely to trigger degradation pathways, such as denaturation of the capsid protein and nucleotides, aggregation, hydrolysis and precipitation and adsorption of the vaccine onto the container walls. This mandates the use of buffers as well as functional excipients and other materials in the formulation, such tonicity agents and stabilisers, non-ionic surfactants to prevent adsorption to glass surfaces and cryoprotectants (sucrose, ethanol or cyclodextrins), free-radical oxidation inhibitors and metal chelators (edetate).

Table 2 below summarises the main differences between Janssen-Cilag and AstraZeneca Covid-19 vaccines’ formulation and excipients:

AstraZeneca vaccine Jansen-Cilag vaccine
Name product Vaxzevria (formerly AZD1222) COVID-19 Vaccine Janssen
Active ChAdOx1-S [Recombinant] Ad26. Cov2.S [Recombinant]
Dose & route of administration 0.5 ml; intramuscular

(containing ³ 2.5×108 Inf. Units)

0.5ml ml; intramuscular

(containing ³ 8.3×108 Inf. Units)

Delivery system Replication-deficient, non-encapsulated Chimpanzee adenovirus ChAdx1-S encoding SARS-COV-2 spike (S) glycoprotein

 

Each virion is 80-100nm and contains a single copy of double-stranded DNA

Replication-deficient, non-encapsulated adenovirus type 26 (Ad26) encoding SARS-COV-2 spike (S) glycoprotein

 

Each virion is 80-100nm and contains a single copy of double-stranded DNA

Diluent Water for injections Water for injections
Buffer system L-histidine, L-histidine hydrocholoride monohydrate

pH = 6.6

Citric acid monohydrate, Trisodium citrate dihydrate

pH = 6.0-6.4

Other excipients Magnesium chloride hexahydrate

Polysorbate 80

Ethanol

Sucrose

Sodium chloride

EDTA

HCl

Polysorbate 80

2-hydroxyl propyl β-cyclodextrin

Ethanol

Sodium chloride

Sodium hydroxide

Table 2. Formulation and Excipients used in Janssen-Cilag (Ad26. COV2.S) and AstraZeneca (AZD1222) COVID-19 vaccines

Differences in Storage Requirements

All vaccines (with the exception of a select few) require high quality and robust cold chains to guarantee stability and viability. These conditions are not arbitrary – they are arrived at from extensive stability studies and conditions where the viability of the products is monitored.

As hinted to previously, the mRNA vaccines are especially vulnerable to handling conditions, hence their requirements are particularly elaborate compared with adenovirus vaccines.

Of the two mRNA vaccines, Pfizer’s is the more challenging to handle, requiring shipping and storage in ultra-cold freezers. I was not able to find any studies on storage stability in the public domain on mRNA COVID-19 vaccines, however Onpattro® , a marketed LNP product has a shelf-life of 36 months when stored between 2° and 8 °C. It is possible that in future, these conditions will be updated as more storage stability data emerge.

A summary of the key requirements for the different vaccines is below:

Pfizer BioNTech COVID-19 Vaccine

  • 6 months maximum shelf life when stored in a freezer at -80°C to -60°C
  • 31 days maximum shelf life at 2-8°C after thaw
  • May be stored between 2 to 25°C for 2 hours prior to dilution after removal from the fridge
  • Once diluted may be stored between 2 to 25°C for a further 6 hours
  • Protect from room light and direct sunlight or UV light

Moderna COVID-19 Vaccine

  • 7 months maximum shelf life when stored in a freezer at -25°C to -15°C
  • 30 days maximum shelf life at 2 to 8°C after thaw
  • May be stored between 8 to 25°C for up 12 hours prior to dilution after removal from the fridge
  • Once punctured, the vial must be used within 6 hours
  • Protect from room light and direct sunlight or UV light

AstraZeneca COVID-19 Vaccine

  • 6 months maximum shelf life when is stored in a refrigerator between 2 to 8°C
  • 6 hours maximum shelf life when stored between 2 to 25°C.
  • Once punctured, the vial must be used within 6 hours
  • Must not be frozen
  • Protect from room light and direct sunlight or UV light

Vaccine Janssen-Cilag COVID-19

  • 24 months maximum shelf life when is stored in a freezer between -25 and -15 °C
  • 3 months maximum shelf life when stored between 2 to 8 °C after removal from freezer.
  • Once punctured, the vial must be used within 6 hours
  • Must not be frozen
  • Protect from room light and direct sunlight or UV light

So there you have it. A summary of platforms for Covid-19 vaccines, the formulations and excipients used, and how these influence handling, storage and distribution requirements of Pfizer BioNTech, Moderna, Astra Zeneca and Janssen-Cilag (Johnson & Johnson)’s vaccines.

 

References

  1. Schoenmaker, D. Witzigmann, J.A. Kulkarni, R. Verbeke, G. Kersten, W. Jiskoot, D.J.A. Crommelin, mRNA-lipid nanoparticle COVID-19 vaccines: Structure and stability, International Journal of Pharmaceutics, 601 (2021) 120586.
  2. S. Rosa, D.M.F. Prazeres, A.M. Azevedo, M.P.C. Marques, mRNA vaccines manufacturing: Challenges and bottlenecks, Vaccine, 39 (2021) 2190-2200.
  3. D’Amico, F. Fontana, R. Cheng, H.A. Santos, Development of vaccine formulations: past, present, and future, Drug Delivery and Translational Research, 11 (2021) 353-372.
  4. Mäder, K. Solid lipid nanoparticles, Handbook of Materials for Nanomedicine, Jenny Stanford Publishing 2020, pp. 173-206.
  5. Tatsis, N., Ertl, H.C., Adenoviruses as vaccine vectors, Molecular Therapy, 10 (2004) 616-629.

 

Subscriptions for the European Pharmacopoeia 10th Edition, Supplements 10.6-10.8 now open

2022 subscriptions to the European Pharmacopoeia (Ph. Eur.) are now open, the EDQM announced on 29th April 2021. Purchases can be made directly on the EDQM WebStore.

The 2022 subscription includes three non-cumulative updates (10.6, 10.7 and 10.8) and provides access to the Ph. Eur. online archives until 31 December 2022.

As has been the case in previous instances, two subscription formats are available:

  1. Electronic (bilingual English/French): This allows authorised access to the entire 10th Edition of the Ph. Eur. until 31 December 2022.

For each licence purchased, subscribers can choose their preferred option: individual licence or shared licence.

Individual licence gives a single user access in three ways: online, from any computer; offline, by downloading it onto one computer or mobile access by downloading it to one USB stick

Shared licence provides access from one shared computer or from one USB stick.

  1. Print (Hard copy)

This is available in English or French (EDQM permits translations into other languages by the member states themselves).

Expect to pay Euro 540 for a single licence (electronic) and Euro 600 (includes a Euro 60 handling fee) for the hardcopy. Special prices are available for multiple and unlimited electronic versions and packages including both formats.

The first edition of the European Pharmacopoeia was published in 1969. A new edition of the European Pharmacopoeia is now published every three years and has been made available in print and electronic for over a decade now.

 

 

NASA’s Ingenuity helicopter

How does NASA’s Ingenuity helicopter flight benefit lifesciences?

NASA’s Ingenuity helicopter made history by flying on Mars on April 19. At 10:49 am (PDT), the tiny helicopter spun its carbon fibre rotor blades and lifted itself into the thin Martian air, rising to a height of about three meters above the ground, before settling back down to the ground. Since this ground breaking flight, a further three flights have been completed, with the latest one taking place on 30th April.

But given the huge costs of this mission (and many others like it planned), many have been asking what the benefits to humanity are and whether or not the huge sums used to fund these programmes could be better spent on more urgent needs?

Ingenuity’s flight is the culmination of more than seven years of conceptualisation, building, testing and hope by the flight team. originally scheduled for April 11, the flight has to be delayed to permit an update of the helicopter’s software. A high-speed spin test on April 16 showed the change would work, setting the stage for the April 19 flight.

With the successful completion of test flights, NASA has indicated it will be scheduling in more tests to figure out the full operational capabilities of the chopper.

“The Ingenuity technology demonstration has been a resounding success,” associate administrator for the NASA Science Mission Directorate Thomas Zurbuchen said in a statement Friday.

“Since Ingenuity remains in excellent health, we plan to use it to benefit future aerial platforms while prioritizing and moving forward with the Perseverance rover team’s near-term science goals,” he added.

While NASA’s Perseverance rover, which successfully landed on February 18, has a clear goal, which is to study rocks and soil samples found on the planet to establish if previous life existed there billions of years ago, the objective of Ingenuity helicopter are not clear cut.

Originally, the Ingenuity helicopter’s task was to show its flying ability, given the differences in atmospheric pressure and gravity found on the planet compared to Earth. The air’s density of the red plan is only about 1 percent that at sea level on Earth. This makes it incredibly difficult for the helicopter’s blades to push against that thin air hard enough to get off the ground. The success of these test flights proves that powered flight is possible in Mars’ thin atmosphere.

Which is all well and good. But what does it mean for those living on Earth right now? The contention being that space exploration of this scale is unnecessary given the humanitarian problems, from COVID 19, the environment catastrophe, hunger and malaria that Earth is facing in this current moment? Perhaps these huge sums could be better spent to solve these humanitarian problems.

However, this argument fails to address the fact that the research and development that goes into these massive projects inevitably benefits Earth, directly and indirectly. True, we may not be able to visualise right now the direct healthcare benefits of Ingenuity helicopter’s ‘wow’ moments but make no mistake, there is much behind-the-scenes preparation and research into materials and software that has gone into this program that is bound to trickle back and benefit us all.

If in doubt, just think of the knowhow that previous space exploration has generated which has in turn been applied to healthcare and lifesciences, from digital imaging technologies, prosthetic limbs, heart pumps, cochlear implants, freeze drying, HACCP and water purification. NASA played a large part in their research and development.

The importance of sustainability to the pharmaceutical industry

Earlier this year, Corporate Knights, a Toronto-based sustainability performance research and media company, released their Global 100 Most Sustainable Corporations rankings. The annual ranking is based on an assessment of more than 8 000 large global companies with revenues > US$ 1 billion. You can obtain more information via this link.

As one would have expected, renewable energy companies dominate, with Ørsted and Schneider Electric, bagging second and first spots this year. Pharmaceutical companies feature on the list, albeit at number 16 (Eisai), 65 (Sanofi), 71 (Takeda), 82 (AstraZeneca) and at 98 (Novo Nordisk).

The fact that pharmaceutical companies feature at all is something to welcome but at the same time the fact that so few pharmaceutical companies feature in the top 100 is disappointing.

Given how processes for the development, production, distribution and disposal of drug products use huge amount of natural, human and economic resources, a lack of interest in sustainability is potentially problematic and only helps further dents the sector’s image as well as its future sustainability.

With society increasingly placing high expectations on corporate entities, it is no longer enough to simply have good intentions, actions must follow words.

But first, what is sustainability?

Sustainability is one of those things that carry different meanings to different people. Sustainability experts say it is simply a business strategy that takes into account an organisation’s operations and their social, ecological and economic impact.

The consensus today is that sustainability is actually good business; a focus on sustainability boosts an organization’s top and bottom lines. This is why a recent survey of top executives at Fortune 500 firms revealed that they now consider sustainability a necessity for competitiveness and future survival.

Examining sustainability literature reveals three main pillars: the environmental, the social and, the economic— this is what is also referred to as profits, planet, and people. The idea here is that by actively addressing environmental and social issues companies can contribute to the society’s sustainability while also achieving their own long-term value (profitability, return on capital, etc).

The environmental pillar is what the majority of us are familiar with, thanks in part to Greta Thunberg . It is concerned with the reduction of carbon footprint, water usage, non-decomposable packaging, and wasteful processes, as part of a supply chain. Cleaning up these processes has been shown to be cost-effective, and financially beneficial while also positively impacting the environment at large.

The social pillar is about treating employees and the communities within which the business operates fairly and responsibly. Some of the important aims here include compassion and provision of responsive benefits, such as better maternity and paternity benefits, flexible working, staff development opportunities, etc.

And, finally, the economic pillar is about running businesses in an economically sustainable way; to be profitable and produce enough revenues long-term. It is not about making money at any cost, rather companies should aim to generate profit in accordance with other elements of sustainability.

Embracing these three pillars, namely social, environmental and economic sustainability is what economists refer to as the Triple Bottom Line.

There is currently no official universal measurement of sustainability in existence, and instead, organisations have developed industry-specific tools and practices to judge how social, environmental and economic principles function as part of a company.

 

Some success stories in Pharmaceuticals sector

AstraZeneca, Eisai, Biogen, Glaxo and Novo Nordisk are pharmaceutical companies that have both worked toward energy efficiency, waste reduction, and other ecological measures. They have also focused on social impact via partner initiatives in the areas of health and safety.

Across the industry as a whole, there is a major shift in thinking, with many companies imposing targets or starting initiatives aimed at reducing the impact of their activities and products on the environment.

Many are exploring ways to produce their products more efficiently and in a sustainable way; such as implementing ‘green’ IT practices designed to lower energy consumption; plastic neutrality and water sustainability.

 

But there’s still a lot to do

As mentioned earlier, the development, production, distribution, use and disposal of drug products has a major impact on environment. For instance, drugs taken by humans and animals find their way into rivers, lakes and even drinking water, and can devastate both aquatic ecosystems.

Sustainability needs to be a priority for any business operating in the sector. And increasingly, people of all walks of life are demanding for it. Very soon, companies will be called to account for all their operations, from carbon footprint, harmful emissions, water usage, etcetera.

The expectation today is that resources should be used responsibly, and where possible, reused to suit the global increase in population.

How do we move from here?

Obviously, there is no “one right solution” on sustainability. The best solution depends on the ambitions and stakes at each company.

Sustainability experts recommend a few useful actions outlined below:

1] Strategic commitment to sustainability: Corporate and business strategies need to be aligned with sustainability.

2] Compliance: There has to be a commitment to comply both with the spirit and letter of the law as it relates to waste management, pollution and energy efficiency.

3] Proactiv response: Rather than wait for a crisis, companies need to develop sustainability strategies today.

5] Transparency: Transparency is pre-condition for measuring and improving sustainability practices. Therefore, companies need to openly communicate with all key stakeholders, openly and truthfully, acknowledging their failures as well as their successes.

To conclude, sustainability remains a major challenge, a challenge that no single company can address. Although the pharmaceutical sector has a lot to do, it is encouraging to see a number of companies embrace sustainability policies. Sustainability is a megatrend that won’t be going away anytime soon.

Reproducing taste with a Norimaki Synthesizer

A Meiji University scientist has invented a method for digitally reproducing taste and flavour in the same just as we do for sound.

 

In the pharmaceutical industry, taste masking of bitter products intended for oral administration is often undertaken during the product formulation stage. The effectiveness of taste-masking methods

Since flavour is a dynamic sense that is influenced by formulation ingredient, effective assessment of taste masking requires actual sensory analysis to be done on the drug product in its final formulation. If the drug product is potent or cytotoxic then sensory analysis may not be possible.

An alternative approach is the use of an electronic tongue – which relies on converting molecular information in a product to generate a signature that can be related to certain qualities of the product, for example, acidic, bitter, salty, etc. nevertheless, the electronic tongue has disadvantages, notably, the need for a huge amount of previous measurements for calibration.

Thanks to Dr. Homei Miyashita, a professor in the aptly named Department of Frontier Media Science, Meiji University, Japan, formulators of drug products will be soon be able to accurately taste and profile their formulations without needing to sniff or taste any product.

Dr. Miyashita interest in food and taste goes a long way back to when he was a child. He has undertaken research on technology and the human senses while at Meiji University, culminating in the Norimaki Synthesizer.

You can read more about Dr. Miyashita’s device through this link: https://meiji.elsevierpure.com/en/publications/taste-display-that-reproduces-tastes-measured-by-a-taste-sensor

The Future of Science Events Post-COVID?

What’s the Future of Science Events Post-COVID?

It’s a cliché, I know, but COVID-19 has had an impact like nothing before on the events industry, and scientific conferences have not been spared. The arrival of the pandemic put brakes on all substantive meetings and conferences, and save for a few hybrid events here and there, it has not been the same since March 2021. Fifteen months on we are still some way before some semblance of normalcy returns when venues and public gatherings will be allowed to operate at full capacity.

Why Events Still Matter

A major activity in the life of a scientist, regardless of the role or sector, is the professional event, such as conference or meeting. Events provide an important space for practitioners to present their work, gain new skills, and acquire fresh ideas that can help further their research/careers.

Events are also important for brainstorming, networking and making vital connections with other scientists and suppliers and can lead to new initiatives, papers and funding. This is why scientific events matter.

From an economic viewpoint, events are a global force, contributing $1.2 trillion to the global economy through direct spend by delegates, attendees and organisers, as well as the millions of jobs supported, directly and indirectly.

The most popular scientific events, ranked by aggregated attendance, are seminars and corporate events, conferences and exhibitions, trade shows and fairs, and product launches. Interphex (mainly US-based), CPhI (Europe, US and Asia) and AAPS (US-based) represent the most prominent events and meetings annually on the pharmaceutical events calendar.

A Change to the Hybrid Model

With the pandemic raging, travel halted, budgets tightened and venues ordered to close, the events sector was forced to rethink and remodel their operations. Smart event organizers saw virtual events as an alternative, opening up opportunities that had never existed in the events industry before. Organisers had a lot of trial and testing to do in order to transition online. And people were excited by the idea of attending virtually as the “new normal”.

Virtual events offered a much needed break for organisers and permitted meetings to continue. As the situation improved, some organisers moved to hybrid events, which allowed some delegates to meet in person whilst also communicating with others virtually.

The hybrid innovation has allowed companies to continue to offer and run events much more frequently, and so far, this does not look like a temporary shift.

And although the number of physical attendees is restricted, hybridization allows extension of the event’s reach geographically, making it more cost effective and a more inclusive experience for both planners and attendees, a real positive. Moreover, as the content can be recorded it can be reused over and over again, increasing its life span.

Hybridisation represents a fundamental change in how events will work in the future, and hopefully, now means even greater engagement by companies and attendees.

The only problem, though, is that virtual events are cutting out most of the supply chain. Suddenly, venues are no longer in as high a demand as was before. Neither are caterers, furniture rentals, sound and lighting engineers, cleaners, and exhibition stand builders. All of these roles are being scaled back with the increasing resort to virtual events.

What Returns and What Changes?

There is no doubt that physical events will bounce back. Science is after all a social endeavour, and as scientists we thrive through physical interaction with other scientists and other humans. Getting back to normal, however, will take a while, and during this time, scientific meetings and events will more likely look very different. Expect more sophisticated virtual participation options, smaller satellite events complementing larger gatherings, and targeted events as people carefully weigh benefits and risks of attending events.

This year, and probably most of 2022, travel will likely remain complicated. But as more positive shoots of economic recovery emerge and bloom, the sector will bounce back strongly. Below is how events professionals predict the industry developing:

Annual general meetings and conferences

The annual general meeting and conference has been and will still remain the main platform for meeting, networking and exchange. Pre-pandemic, the annual meeting and conference typically attracted 500+ attendees, convening centrally, and interacting closely. But in the age of Covid-19, conferences do not sit well with social distancing requirements. This has made them nearly impossible to hold – both logistically, due to travel restrictions, and as a matter of public health.

When they return, the annual conference and meeting will likely look very different. You can expect shorter, more sophisticated events, with more virtual participation options, greater use of event technology, mobile apps and social media. We will likely see event split between the main event (50 to 100 attendees) and many more break-out, intimate sessions of anywhere between 10 – 15 attendees.

Satellite Events

With mass meetings being one of the proven vehicles of spreading COVID-19, it is understandable why people will be sceptical about attending live conferences soon. Therefore, smaller events with a manageable audience are the immediate future of the industry. You can expect to see smaller satellite events aimed at complementing larger gatherings. So if the main event is in London, you might have smaller hubs in Frankfurt, and Madrid.

Targeted Events

For a long time now, many organisations have been questioning the ROI on tradeshows and conferences. So while interest in targeted events, such as roadshows, trunk shows and townhall meetings, had been growing, we will see interest in these grow. The advantages of roadshows is that they can either be stand-alone events or a series of identical meetings taking place sequentially in multiple locations and allow companies to take their message out and meet targeted audiences face to face. Think of it as the event going to where the audience is rather than the audience coming to the event.

Event Technologies

Event technology is a term used to describe all of the digital tools, apps and software used in the events industry. Everything from check in & registration, to diagramming, to social media tools, and more can be considered event technology.

Acceptance of event technologies has been increasing over time and we now should expect event organisers and planners to increasingly leverage technology not only to manage COVID-19 risks but also to improve attendee experiences.

Takeaways

The pandemic has forced the event industry to adapt to various challenges at an unprecedented speed. This has even pushed event planners to innovate, as exemplified by increasing adoption of hybrid and virtual eventing.

Although virtual events bring the benefits of removing time and space constraints and increase participation, the need for social interaction cannot be dismissed.

Going forward, we can be sure to see changes in the way scientific events are delivered, including greater use of technologies, shorter, more intimate physical events and many other as yet to be implemented innovations.

Back to Top
Product has been added to your cart