Tuesday, January 16, 2024

Biopharmaceuticals And Medical Devices: A General Overview II (Drugs)

 In Biopharmaceuticals And Medical Devices: A General Overview I, we looked at the underlying basis of all drugs (pharmaceuticals and biologics) and discussed the development of a compound from initial discovery to the completion of Non-Clinical Laboratory Studies and the decision to move forward.  Today, we are picking up with starting clinical manufacture and clinical trials.

There are really three streams here:  clinical trials, clinical manufacture, and regulatory submission.  I am going to defer the regulatory submissions to later in the week as there are some small similarities with the Medical Device process.

Clinical Manufacture

Clinical manufacture is the manufacture of a drug to support a clinical trial.

Typically, clinical manufacture is divided into two phases, the manufacture of the actual compound (called an "Active Pharmaceutical Ingredient" for pharmaceuticals and "Drug Substance" for biologics) and the manufacture of the finished form of the product such as pills, capsules, filled liquid vials, lyophilized solutions, etc. called the  Drug Product.

The work done in the investigatory and pilot plant manufacture will be scaled up in volume to support the clinical trial, for example from a 5 L fermentation to a 50 L fermentation to a 600 L or 1000 L fermentation.  Process Development will work to find the ideal conditions for manufacture and all required buffers, medias, or reagents.  Process Sciences will develop criteria to evaluate the product - called "specifications" - which will allow testing during manufacture ("in process"), after manufacturing ("release"), or over time to evaluate how long the drug product remains "good" ("stability").  They will also identify tests to support these criteria.  

At some point, the developed process and associated tests and specifications will be transferred to the Operations Group.  Here, a variety of operations - Manufacturing, Quality Assurance, Purchasing and Supply Chain, Quality Control - will purchase and test incoming raw materials, write and approve manufacturing records for every part of the process, write and approve testing documents for all identified tests, and create final release documents so the product can be evaluated and made available for clinical trials.  The facility to be used will have to demonstrate that the meet the required regulatory and physical conditions for manufacture including temperature and humidity control, air particulate control, and micro-organism control. The equipment to be used - both manufacturing and testing - will need to be qualified to verify it is installed correctly, works correctly, and works for the process. Likewise, all quality control testing will have to be qualified to verify that the tests are repeatable, give verified data, and can be performed by multiple personnel potentially in different labs.

One note: development of the manufacturing process and manufacturing may be conducted in-house at the company or outsourced to a third party vendor, called a Contract Manufacturing Organization ("CMO") or Contract Development and Manufacturing Organization ("CDMO").  There are trade-offs between time and money; for many small companies they have no choice but to use a CMO as building out and qualifying an in-house manufacturing plant can be expensive and time consuming.

In the background of all of this (and by regulation), every company has a Quality System, which is the System put in place to guarantee that all product manufacture is conducted in such a fashion to ensure the identity, strength, purity, quality, and safety of the product.  This includes instructions on how to do things ("Standard Operating Procedures", or SOPs) for every aspect of the organization that is regulated, how to evaluate issues and failures (Deviations or Nonconformities), how to make improvements (Corrective Actions and Preventive Actions), how to manufacture and test the product (as above), and how to release the product. For all of these documents, evidence of training must exist.

Assuming all of this is in place, manufacturing will start.

Manufacturing will often start with a "non-GMP" batch, sometimes called an Engineering Batch.  This lot cannot be used for clinical trials but can demonstrate that the process can generate the Drug API/Drug Substance/Finished Product at sufficient scale and with sufficient quality.  If successful, manufacturing will move into GMP/ Clinical Trial material mode.

As trials advance (see below) the manufacturing and testing will progress as well.  As more is known about the product, tighter controls will need to be put in place.  Sometimes a problem only reveals itself over time through stability, which may mean going back to development.  Better and more extensive testing will develop.  And the process size will scale up as well to support commercial manufacture; for biological products, up to 20,000 L fermentation vessels exist for manufacturing

Clinical Trials

Clinical trials for Drugs in the U.S. (and largely, the world) are divided into three phases.  Each phase gathers information about the product and its impact on patients, but each phase also has a primary goal:

Phase 1 (also Phase I or P1) evaluates safety, or "Is the product (at a high level) safe?"

Phase 2 (also Phase II or P2) evaluates efficacy, or "Does the product do what we think it should do?"

Phase 3 (also Phase III or P3) evaluates dosing range, or "What is the best dosing level that maximizes benefit and minimizes patient impact?"

The primary concern of all Clinical Trials is ultimately the safety of the patient (this largely derives from World War II, where live in human testing was conducted (Read up on the testing in concentration camps and Unit 731 of the Imperial Japanese Army.  It is horrifying.).  To this end, 21 CFR Part include items specifically around the involvement of patients, physicians, and companies (Parts 50, 54, 56, and 312) as well as the International Committee on Harmonization document E6.  In short, patients must be fully informed and able to withdraw at any time.  Physicians cannot have a significant financial interest in a company for which they are testing a Drug Product.  An Independent Review Board ("IRB") must review all documents associated with a trial and make sure it protects patients, is ethical, and is scientifically justified.

To use an unapproved Drug Product ("investigational drug" in the regulated world) in humans, the company ("Sponsor")  is governed by 21 CFR Part 21 Part 312, Investigational New Drug Application.  In short, one has to provide an Investigational New Drug Application (called an "IND") and have it approved by the US FDA prior to starting the trial.

The IND will provide a description of the product, a history of its development, proposed release and stability criteria,  a summary of the Non-Clinical Laboratory Studies (GLP and non-GLP Compliant), and the proposed Clinical documentation.

Leading up to the IND, the company is developing documentation for the Clinical Trial:  an Investigator's Brochure, to explain the product and the trial to physicians and site staff; a Pharmacy Manual to explain how to prepare the product for patient use; an Informed Consent which explains to the patients about the trial and their potential participation and potential risks; a Clinical Protocol, which explains how the trial is to be conducted, what is to be tested, how patients should be included or excluded based on criteria, and the endpoints of the trial; the Statistical Analysis Plan created, which ensures that the study has the minimum of number of patients to power the study statistically and can deliver definitive data; and the Clinical Trial Documentation, which is used to record all data for the trial (often this is now electronic).  All of the documents are reviewed and approved internally by the company, by the IRB and by the FDA upon submission of the IND.  Any of these parties can require changes to any of these documents, which then need re-approval.

At the same time, the company is also identifying sites which would be willing to conduct the clinical trial. This is a lot more work that it may sound like:  there are always more trials than sites and some sites are more desirable than others.  For each site a contract must be written and signed, a budget prepared and approved (sites are paid to conduct these trials), all the documentation above provided, and supporting supplies provided.

Conduct of the Clinical Trial

Assuming all the product is manufactured and approved, all clinical documentation is approved, clinical sites have been identified and initiated, and the IND has been approved (or revised and approved as needed), the clinical trial can start.

Clinical trials escalate in size and scope as they progress.  A Phase 1 trial can have as few as 20 patients and 3 sites; a Phase 3 trial can have as many as 7,000 patients and multiple sites in multiple countries.  Study progress is based on the ability to enroll subjects - and have them complete the program.  Not all patients can be evaluated.  Patients may be rare, depending on the inclusion criteria.  If an Unexpected Adverse Event or Serious Adverse Event happens - something that causes transient, partial, or permanent damage to the patient including death or disablement - the trial may be stopped temporarily or permanently. 

During the trial, company personnel from the Clinical, Medical Affairs, Regulatory Affairs, and Quality Assurance departments work intensely with the sites.  Clinical Trial Monitors or Clinical Trial Associates contact and/or visit sites frequently to check up on the trial and audit documentation and product.  Medical Affairs personnel assess data and answer questions.  Quality Assurance evaluates potential complaints.  Regulatory Affairs reports any Unexpected or Serious Adverse events to the Regulatory Authorities.  Meanwhile as needed, Manufacturing, Purchasing and Supply Chain, Quality Assurance, Facilities, and Quality Control continue to do their jobs to ensure that Drug is available for the study.

Periodically the trial may be evaluated by statisticians to evaluate the data.  There may be a reason to cut the trial short, either because the data is overwhelmingly indicative of the endpoints or the data demonstrates no progress to the endpoints or is even risky.

Thoughts

As you can hopefully see, there is a lot to "testing the product in a person".

Perhaps unsurprisingly, a main factor is cost - not just of the all the work to make the product and get to the clinical trial, but for the clinical trial itself.  I would hesitate to provide a number for even the most basic of clinical trials, but it is easily in the millions of dollars for a Phase 1 trial - and goes up from there.  

And the cost escalates of course.  Phase 2 trials are more expensive than Phase 1 trials, and Phase 3 trials are even more expensive.  And the cost of scaling up manufacturing and testing increases as well.  

The funding of a company really determines its future at these stage. Most companies have enough funding to complete a Phase 1 clinical trial.  The hope is that the data from that Phase 1 trial will support getting additional funding to continue to Phase 2, then Phase 3.  Obviously, a failed Phase 1 trial pretty much can spell doom for an early stage company and many layoffs occur after a "setback" in the Phase 1 trial.

But it can happen at any step.  Phase 2 trials fail as well, and Phase 3 also.  The cost of those failures is significantly increased in terms of money as well as prestige:  a Phase 3 trial means that at some point, some signal was missed in an earlier trial.  This can reflect poorly on the company's executive management and scientific basis.  Established companies with commercial products can survive this; most non-commercial companies cannot.

And we have still not arrived at the ability to sell a single Drug Product and thus generate any revenue for all of the work done to date.

Tomorrow we will turn our attention to Medical Devices and how they reach this same point of pre-submission.

11 comments:

  1. When my mom was looking at signing up for several studies as a last resort for her particular brain cancer, I did a lot of research into what you talked about above. Unfortunately, she only qualified for a Phase One test and only received one dose in the end which didn't do anything for her.

    I do have a question. At what point in the process does a medical company begin the patent process? I would assume in today's environment, they begin the paperwork as soon as there is a thought that it might be something of value to pursue. Do they continue to patent the drug even though it might not make it through clinical manufacturing studies or even all three phases of clinical trials? Do they wait until everything has passed to file the final paperwork? I know with drugs, timing is very critical since the patent only lasts for a number of years compared to say a mechanical patent for a machine.

    ReplyDelete
    Replies
    1. Ed, you touch on two points that I intend to touch on in more detail Thursday or Friday.

      1) One very wise person long ago reminded us that most Phase 1 patients, especially in Cancer Trials, are essentially self-sacrificing volunteers. For most, they have already gone through standard treatment assessment and options and their chances of seeing results in any clinical trial, especially an early one, are very slim. Essentially, they sacrifice their hope of survival to advance knowledge.

      2) Intellectual Property (IP) was something that I thought about touching on in today's post (but then, obviously, got carried away). The short version is there are two elements. The first is doing a search to make sure no-one else has the patent on a construct (it has happened before at I place I worked and money was spent on a compound that could never be used). The second is that they have to have enough information on the construct to patent it (for example, one could not patent an entire class of ACE or BRAF constructs; there needs to be a bit more information than that.

      Likely once a patent is filed it will be held onto - who knows if it can be sold or will be valuable later (see "the little blue pill" Viagra here; initially developed for vascular expansion, it was found to have "other effects").

      It is also important to differentiate between a patent and a drug approval. A patent (per Wikipedia) runs 20 years currently. A Drug Approval has varying terms of exclusivity based on the type of product and the target market. A (pharmaceutical) appears to currently be five years, but if it for some conditions (such as so-called "orphan drugs", where there are small populations) the exclusivity can be extended to incentivize the development of the product. Biologics are currently 12 years, probably partially based on the fact that a biologic is a more complex item to develop and manufacture (as they are created in living systems).

      To be clear, I am not a lawyer and there are undoubtedly permutations I am not aware of. But at a high level, these are relatively true.

      To answer your initial question, effectively the clock is ticking the moment the patent is filed so there is a balance between filing and investing time and effort appropriately.

      Delete
  2. Nylon125:25 AM

    Providing this information on your industry is quite interesting TB, looking forward to the rest of your submissions.

    ReplyDelete
    Replies
    1. Thanks Nylon12.

      Honestly, this has been good for me as well. 95% of this is pulled directly from brain (even I do not remember everything), so the fact that I have gained so much knowledge over time is strangely comforting.

      Delete
  3. Given the multiplication of the human effort using computers. And the Loss of many jobs replaced by those computers.

    What effect on drug and device biomedicals development that AI will have. Already I've been reading in Barrons and such the cost savings and accelerated development in those lovely gene vaccines and such.

    ReplyDelete
    Replies
    1. On one hand, AI will indeed impact the industry. On the other hand, getting to potential compounds faster means that those compounds can make it to the lab for further investigation and research.

      There is cost savings and personnel savings which is likely contracting parts of the industry. On the other hands, things like biostatistics are booming. And automation has been a thing for some years, especially in drug product manufacture. Lines to fill liquid vials, the part I am most familiar with, can be a pretty impressive demonstration with very limited human intervention.

      Delete
  4. Getting caught up on this interesting series. Thanks for tackling it, TB. As usual, it's well researched and well written.

    I recall some of what you're talking about from nursing continuing education courses and it all makes sense on some level. Even so, I now wonder about some of the things Michael mentioned, especially the fast-tracking we've seen in the past several years, which enables the sidestepping of much of the testing. I thought that was supposed to be a one-time emergency proposition, but it seems to be becoming a standard of it's own.

    Then too, we've seen an across-the-broad generalized shift in ethical standards in recent years: top-heavy executive salaries and bonuses, non-transparency of drug and product mark-ups, recruiting physicians and hospitals as sales affiliates for drug sales.

    Another problem I used to see when I worked in the hospitals was a general disregard for waste amongst the bottom-rung employees. The attitude was, 'so what, insurance is paying for it anyway.' While I strongly disagree with this attitude, I couldn't blame them for because we all had heavy patient and work loads and no one had the time to care.

    Back in those days, the public conversation was "the rising cost of health care." But Obama shifted the conversation to "affordable health care." The problem I've always had with that is - who defines "affordable?" It's vastly different for our wealthy political betters and those of us trying to live a half-way decent life, take our kids to McDonalds once in awhile, and put a little aside for retirement and future needs.

    As with all things, there is an official viewpoint, and then there's reality. If anything taught me this, it was nursing. Then, when Dan cut off his finger several years ago and we had no job, no income, no insurance, and no advocate, we saw a side of it all that most people don't see. Can't say I have much hope for the whole thing.

    ReplyDelete
  5. Part I:

    Leigh - Glad it is turning out to be of use (or, as the saying goes, "Be careful what you ask for...").

    A note on the testing that Michael referred to: AI and computers in general have speeded up the initial identification and early research process (the "R" in R and D). Clinical Trials - the In Vivo part - are still required. The US FDA is allowed to reduce that testing under the Emergency Use Authorization or EUA (21 CFR Part 312.36), such as was done during The Plague. Two thoughts there: the first is that this is a real risk in that the long term effects and sometimes the short term effects will not be known. The other is that if it is used too many times, it loses the benefit: people are smart and will figure out the riskiness of the proposition.

    I am always a bit ambivalent about the question of salaries. Perhaps an example from my recent employer will suffice: I was paid a fairly small amount higher than my senior direct report, yet I ultimately held the responsibility for making Quality decisions. To be completely frank, it was not enough for the responsibility. If I decided wrong, I would get blamed for a bad decision (and in this industry, potential injury or death). Was 7% more really worth that? Probably not. I will not argue the amounts, but I will argue that if you do not pay according to responsibility and risk, you will eventually get either no-one willing to make a decision or people that should not be making them making them.

    Costs are a bit tricky as well, both from the total amount as well as for the individual components. For example, people are always the biggest expense of any operation. One easy way to lower costs anywhere is to pay people less, which in a way defeats other purposes. Material costs are another one that have an impact and have increased significantly as well as their availability: for example from 2020 to 2023 plastic tubing was simply not available or on backorder for months on end - or you could pay a premium from somewhere else to purchase it.

    I suppose one could make the argument of a "fixed percentage increase" - but like Social Security, I suspect companies would rapidly find it was not actually meeting the true costs and would then make tough choices. It does happen know sometimes: companies wind down production because there is simply not enough profit to make it worth their while. I always imagine what it would be like for those dependent on such medications to hear that they could no longer product them due to cost. Could the government force them to? Probably. But likely such production will get the minimum of attention and effort.

    "Affordable health care" is one of those concepts like "living wage": it is definitely in the eye of the beholder. For example, I can get generic versions of many common drugs for $10 to $20 a prescription. I assure you that is not the cost of manufacture. It is already being subsidized, mostly through insurance fees and the government. So if I am paying taxes on something to make it affordable or livable for myself or others, am I really enjoying that affordability or livability or is it simply another tax I pay?

    ReplyDelete
    Replies
    1. Part II:

      One of the core points you bring up is people not caring about how money is spent. This is not just true in health care, it is true in virtually every area of the U.S. economy. People by and large do not treat money as their own and thus, feel no responsibility towards it. Nothing is truly free: it is always paid for by someone. If people perhaps realized that by wasting they are actually increasing their costs overall, they might do things differently.

      I do remember Dan's finger and your experiences. And I, too, am not hopeful. I know that - for the healthcare side - many people suggest some version of socialized health care. Yet those that suggest it always only talk about the positive sides, mostly the universal coverage and low out of pocket expenses. What they do not discuss are lengths of time to get things like appointments and procedures, the disincentives for improvements and innovation, and the fact that the low out of pocket expenses are subsidized by high tax rates.

      Bottom line, and something I have seen elsewhere and believe: more than ever, we are responsible for our health. It strikes me as odd in this innovate and technological age, but there it is.

      Delete
  6. The advantage of me coming along and reading these interesting posts is that the comments are filled with even more cool stuff. This is a great series, TB. The idea of clinical trials kind of fascinated me while I was in the midst of diagnosis and treatment last year. I was curious enough that I went online to see if there were any trials I might qualify for (somehow I had a link - might have been provided by the oncology center I was/am a patient with). I didn't appear to qualify for anything, but I couldn't help but think doing a trial would add some interest to and greater purpose to what I needed to go through. I appreciate the glimpse inside.

    ReplyDelete
    Replies
    1. I really do have one of the best Commentariats on the InterWeb.

      Your physician might have provided you with a list - the Federal government maintains a list of all clinical trials (https://clinicaltrials.gov/). To your point, they often have a pretty specific set of requirements to participate in the trials (called inclusion/exclusion criteria) which incorporates things like nature of the disease, health, previous drugs taken etc.

      To my point to Ed above, participants are often the unsung heroes of moving the ball forward, especially in diseases like cancer. The chances of a successful result in such a trial is generally small (especially in the early days), but the information gained can help the entire field in terms of what we learn about the disease.

      Delete

Comments are welcome (and necessary, for good conversation). If you could take the time to be kind and not practice profanity, it would be appreciated. Thanks for posting!