Showing posts with label biosimilars. Show all posts
Showing posts with label biosimilars. Show all posts

Thursday, May 9, 2013

Bringing In The Big Guns

In the biotech world, perception can be (and often is) reality. The perception that you brought in the big guns matters as much as the reality that you brought bullets.

It actually reminds me of that scene from the movie, Men In Black where Tommy Lee Jones' character is arming Will Smith's character with this puny gun:



In the movie, this gun is called the "Noisy Cricket" and turns out to pack an enormous punch. However, the gun's small sizes makes it look like, at best,  a weak weapon.  Look at Will Smith's face.

What does this have to do with bioreactor sterility or cell culture consulting?

Well when you have a rash of contaminations or when your production campaign is on track for failing to meet ATP, you have actually TWO problems:
  1. Solving the actual problem
  2. Looking like you're solving the problem
A customer once told me, "Oliver, I need both action and PERCEIVED action."

He's right, and here's why:

In claiming to have solved a problem, the first thing people will usually ask is how you solved the problem. If you tell them the solution and the solution is credible (perceived to be viable), then great, problem solved and let's move on.

If you tell them the solution and the solution seems dubious even though it worked, you will get lingering questions and second-guesses.

Managing the perception is as important as managing the reality.

Sunday, May 5, 2013

Amgen's Biosimilars Offensive

Here's an article at Genetic Engineering & Biotechnology News on the top 10 innovator drugs competitors are trying to copy.
  1. Amgen's Aranesp
  2. Amgen's Enbrel
  3. Amgen's Epogen/Procrit
  4. Pfizer's Genotropin
  5. Roche's Herceptin
  6. AbbVie's Humira
  7. Amgen's Neulasta
  8. Amgen's Neupogen
  9. Janssen's Remicade
  10. Roche's Rituxan
This list appears to be alphabetical (by the drug's trade name).  As you can see, 5 of the top 10 biologics that are marching inexorably towards patent expiration belong to Amgen.

amgen logo It's no wonder why Amgen needs to start playing offense.  And it's no wonder why they need to start now when the regulatory pathway for biosimilars is still a work-in-progress (WIP).

I'd like to see these companies get more lean and compete on manufacturing agility as well as efficiency.

Processes need to be better specified... the operating space ought to be better characterized (QbD).

Once the technology is transferred to large-scale, there ought to be a program to continuously improve these processes to eliminate variability and increase process understanding (PAT).

All of this manufacturing sciences and technology is already happening, but I have yet to see companies trot it out as a core competency.

It's not sexy, but in a world where your top-line revenues are in decline, focusing on bottom-line numbers is one of many viable paths.

Friday, March 15, 2013

Antibodies (mAbs) were 5 of top 8 best selling biologics

According to @CellCultureDish, 8 of 20 top selling drugs from last year were biologics.

#1. Humira (adalimumab)
#2. Remicade (infliximab)
#3. Enbrel (etanercept)
#4. ~not biologic~
#5. Rituxan (rituximab)
#6. Lantus (insulin glargine)
#7. Herceptin (trastuzumab)
#8.~not biologic~
#9. Avastin (bevacizumab)
#10.~not biologic~
#11.~not biologic~
#12. Neulasta (pegfilgrastim)

Source: Genetic Engineering and Biotechnology News

It's also interesting to point out that 5 of the 8 are monoclonal antibodies (mAbs). You can easily tell from the chemical name of the biologic because the chemical name ends with -mab.

Anatomy of the Antibody

For the non-biogeeks out there, monoclonal antibodies are Y-shaped molecules that are naturally produced by our immune systems to fight off foreign germs. Picture doing the "Y.M.C.A" dance and you're doing the "Y".

YMCA village people
The Village People emulating the shape of a monoclonal antibody

The significance of antibodies is that they are highly specific, meaning that they will bind to one target and only that target; it's possible because the antibody's molecular "hands" fit only one "glove" (specific antigen).

So the reason why monoclonal antibodies are so useful in medicine is because they can hit the targets that you want and nothing else... so-called, "Smart-bombs." And you'll see a lot of mAbs in cancer treatment (since you want to target only the cancerous cells, but not the healthy cells).


Equivalent of when an antibody comes across an antigen it doesn't recognize

mAbs can be divided into two regions: Fab and Fc.

Fab stands for fragment (antigen-binding). The part of the antibody that binds to the antigen. The Fab is basically everything from your shoulders up.

Fc stands for fragment constant: the rest of your body from your shoulders down to your legs. It's not really constant as it can be one of several classes in humans; but relatively, it's constant.

Early antibodies were engineered from mice. In very simplistic terms, you introduce a foreign biological molecule (antigen) to a mouse. The mouse's immune system will naturally fight off this foreign molecule by producing antibodies, and voila, you have yourself a mAb that can target your antigen. This mAb is 100% murine.

The problem with murine antibodies as medicine is that the human immune system recognizes it as foreign and will reject it. So to be useful, the antibody needs to be "humanized."

Humanizing an antibody means replacing mouse-Fc region with a human Fc region and then trying to make as much of the Fab region as human as possible.

mAb Nomenclature

There's actually a nomenclature for monoclonal antibodies that can tell you from the name, how much of the antibody is murine and how much is human:

diagram mab antibody

In the above diagram, the light-blue antibody is from a 100% murine (-omab). The darkish-red antibody is 100% human (-umab).

The genetically engineered antibodies are the ones that are mixed in color. A chimeric antibody (-ximab) is one where from the "elbow" down is human, and from the "elbow" to the fingertips is murine. Drugs like Remicade (infliximab) and Rituxan (rituximab) are chimeric.

A humanized-antibody (-zumab) is one where only the "fingers" are murine and the rest is human. Drugs like Herceptin (trastuzumab) and Avastin (bevacizumab) are humanized antibodies.

While the more humanized an antibody is, the less rejection it gets as a viable drug, there appears to be no obvious correlation from a revenue perspective (see list above).

What's this have to do with cell culture? Antibodies are secreted by genetically-engineered cells that are grown in cell cultures.

Related posts:

Follow me @zymergi






Tuesday, March 5, 2013

The Crux of Biologics Manufacturing

Whether you are making a biologic or a biosimilar, the manufacturing process (for the most part) is the same.

You're going to have cell culture or fermentation in either as a batch or continuous process.

You're going to have a recovery/purification train of 3 or more steps where you're purifying the drug and getting rid of unwanted biochemicals.

You're going to freeze that into huge ice cube and ship it to your fill/finish facility where they're going to thaw it and dispense the drug product into vials.

What biologic you make and how hard it is to make was decided by your Process Development group years ago when they were poking the DNA into cells, growing them and picking the "best" one. (This is a big-time simplification, the wonkish version of expression system selection, cell line development, cell line engineering, platform development, clone selection, process characterization/validation... etc.)

What decides whether you're making bevacizumab or rituximab?

If you inoculated a bioreactor with the cells that were transfected with the anti-VEGF gene, you're going to get bevacizumab.

If you inoculated a bioreactor with the cells that were transfected with the anti-CD20 gene, you're going to get rituximab.

It goes without saying that you must follow the recipe/manufacturing formula for the respective cell lines, which will contain minor differences across upstream and downstream.

The crux of biologics manufacturing is cell line development and what makes biologics manufacturing hard was doing the cell line development right.

What decides whether you're making Rituxan® or Reditux®?

If you inoculated your bioreactor with cells that were transfected with the anti-CD20 gene by an IDEC scientist, you're going to get the biologic: Rituxan®.

If you inoculated your bioreactor with cells that were transfected with the anti-CD20 gene by a Dr. Reddy's scientist, you're going to get the biosimilar: Reditux®.

What no one knew a decade ago (2003) was whether or not the Dr. Reddy's cell line could produce a molecule identical to the IDEC molecule. And even if they could, would this molecule be as safe and as effective as the FDA-approved IDEC molecule?

Is Making Biologics Hard?

This is a very broad question. Is putting cells into a bioreactor and watching them grow hard? ...because that's how the active pharmaceutical ingredients get made.

Is pouring the HCCF down chrom columns and hard? Broadly speaking, I'd say there's no shortage of people who can execute a large-scale biologics manufacturing process.

Is cell line engineering, platform development, clone selection tough...etc hard?  The grunt work can be done by entry-level scientists/engineers, but you probably want Ph.Ds leading the team to ask the right questions and to cast aside the technical road blocks.

Can Amgen take market share from Roche, Lilly, Abbvie and Janssen? Sure. Amgen has the right people, the resources and leadership.

Is making biologics so difficult to make that no one else can do it?  I don't think so. There's already generic versions of Aranesp, Neulasta and Neupogen in ex-US markets.


Tuesday, February 19, 2013

Amgen's Biosimilars Gambit

amgen logoAbout a week ago, Amgen rocked the biotech industry's proverbial boat with their announcement that they'd be entering the biosimilars market. Multiple news outlets like Yahoo!, Forbes, and CNBC report that Amgen, starting in 2017, will be making six generic versions of blockbuster biologics:
This comes as a surprise to many, because for years, Amgen has been saying that biologics really can't be copied.

You see, Amgen is facing patent expiration on it's blockbuster biologics. And unlike pharmaceutical companies, there really wasn't a need to be worried about patents expiring for biologics.

The U.S. Food and Drug Administration has long held that not only must the product meet quality specifications, but also the process that makes the drug must also rigorously meet process specifications.

Well, biologics are made by genetically-engineered microorganisms (cells) that conduct a symphony of biochemical reactions. You give these cells media; you control their environment (temperature, pH, dO2), and the cells do all the work making your protein out of the DNA.

Need to manage your cell culture data?

Since the process specification includes these proprietary cells, it stands to reason that no one can produce the drug product if they don't have your cells. For this reason, biotechnology companies have not been as worried about patent expiration.

Up until this announcement, Amgen has been focusing on defending their superior position by indicating how difficult it is to manufacture biologics and how consumers ought not to trust biosimilars:
"It's really hard to manufacture biologics."
"When copies of our drugs are made, you can't be certain of their safety/efficacy."

But all of the sudden, our ancient weapons dealer who has been selling us his impenetrable shields has a new offering: his spears that can penetrate anything: 自相矛盾.

Amgen sees the writing on the wall. The FDA is being forced to develop a biosimilars approval pathway as a part of Patient Protection and Affordable Care Act (aka, "ObamaCare"). By law, the only way to do this is to renege on the "product and process" cGMP principle.

One of two things is going to happen:
  1. The FDA is going to allow biosimilars into the US markets.
  2. The FDA is NOT going to allow biosimilars into the US market.
Someone at Amgen put their brain on and decided no matter what happens, they were going to win.

Consistency issues aside, I think Amgen's gambit is genius.

Thursday, January 31, 2013

Amgen Singapore : I stand corrected

In a previous post, I indicated that biotech manufacturing facilities in Singapore costing 200M is cheap.

It turns out that that's par for the course.

My original statement is false, but becomes true when edited thusly:

I think the [Lonza-built] Roche plant cost 290 million and the [Genentech-built] Roche plant cost 500 210 million, so if Amgen gets it done for 200 million, that's pretty cheap par for the course.

I got the 290 million figure from multiple sources:
But then I mis-read the $500 million figure from the Roche Singapore About page where it says:
With a combined investment of approximately USD500 million, the site is comprised of two state-of-the-art facilities which use two different production technology platforms to manufacture biologic medicines.
(emphasis mine).

So doing the math.... both facilities cost approximately 500 million USD.  One costs 290M, so the other must cost $210M.

Thanks to a savvy reader for this correction.

Monday, September 24, 2012

@CellCultureDish On Eliminating Animal-Derived Components - ಠ_ಠ

In a recent article on key improvements for improving the quality of biopharmaceutical manufacturing, the author, "The Dish" states:

Many problems with respect to contamination are a result of problems with raw materials and too often these raw materials are sourced from animals. Animal-derived products always carry a risk of contamination from adventitious agents, such as viruses and prions. Not only are animal sourced products a safety risk, but also they simply are no longer necessary ingredients in manufacturing.

While I agree that eliminating animal-derived sources will improve the process, this claim is rather spurious.

Let's go through this one claim at a time:

Animal-derived products always carry a risk of contamination from adventitious agents, such as viruses and prions.

This is certainly true... there is always a risk, albeit small.  Peptones and other animal-derived media components are heat-inactivated.  Automation engineers code recipes into the PLC or DCS to make certain this happens.  After the animal-derived media components are added during cell culture media preparation, the entire media is virally-inactivated with a process similar to pasteurization.  Whatever risks were there are significantly diminished before the bioreactor is even batched.

In the case of antibody manufacture, the Protein A chromatography is the step that binds the antibody and throws everything else away.  To present the risks of viruses or prions in the final product as a risk - given the viral barriers and purification capabilities of modern biotechnology - is extremely irresponsible.

What about this claim?

Not only are animal sourced products a safety risk, but also they simply are no longer necessary ingredients in manufacturing.

Is it true that we fully understand the components of animal-derived ingredients?  I'm not aware that this is true.  Perennially, miniferm experiments confirm that cultures that use animal-derived sources produce higher titers than "veggie" media or fully-defined media.

There's that je ne sai quoi in animal-derived media that the cells seem to like.  Some think it is the shear-reduction capabilities; others think it is some magic ingredient that we have yet to fully characterize, but these process R&D managers aren't stupid.  If your process titers are low, and using animal-derived media can increase titers so that you can commercially manufacture the API... you use animal-derived media.  It's that simple.

All this said, The Dish is correct about eliminating animal-derived media components... but for the wrong reasons.

Elimination of undefined media sources will reduce process variability because you've eliminated a potentially huge source of variability.  I've seen titers swing 300% from year to year with no change in the manufacturing formula, no change in manufacturing execution, no change in anything but the lots of ingredients that we get. Using defined media will simply eliminate the lingering question of, "Is it the peptone lot?"

On top of that, variability reduction increases process capability.  Even if using fully-defined media loses out 25% yield to the best performing undefined media, you gain from the predictability of your process.

Now, you can schedule campaigns with greater confidence.  You can manage the supply chain.  You can be assure that there will be fewer disturbances to your manufacturing process... you can keep less inventory... and you can ultimately save in costs.

Variability reduction is the reason you want to eliminate animal-derived media components... not risk of viral or prion contamination.

Process R&D managers that "get it" understand that optimizing titer must be balanced with optimizing process capability.

Wednesday, September 19, 2012

Single-Use Bioreactors (there's the Mobius by @EMD_Millipore)

The concept of throwing away your bioreactor after a single use is quite foreign to me. My professional career grew up on stainless steel bioreactors that sat majestically in these uber-clean processing space in these plants.

But the disposable bioreactor concept appears to be gaining traction.

The idea is this:

You sterilize a plastic bag and you fill it with media. You can mix the contents of the bag. You can control temperature. You have pH and dO2 sensors hooked into a control system that maintains pH and dO2. You have everything that a stainless steel bioreactor has to maintain cell culture. Except when you're done with the operation, you get to throw away the bioreactor, whip out a new sterile bag and repeat.

What's the Advantage?

The most obvious thing is that you don't need to clean-in-place (CIP) or steam-in-place (SIP) the bioreactor... these bags come sterile. This means that you don't have CIP and SIP validation and the associated quality costs.

This also means that you don't have the piping, the valves, the boilers, the steam traps... all the infrastructure that goes into maintaining CIP and SIP systems.

CIP and SIP consumables don't need to be purchased. WFI systems can be smaller. Your Waste Kill isn't taking on all that acid, alkali and rinse solutions.

Staffing overhead to maintain and troubleshoot these systems don't need to be hired. The PLC logic or DCS recipes don't need to be coded. Control loops don't need to be tuned, so your automation engineers aren't as busy.

What about contaminations? Everyone knows from miniferm experiments that tube welding has low associated contamination risks. Everyone know that your failure rates go up the larger your tank. Is it true that success rates are higher with these single-use bioreactors?

What Stays The Same?

You're still going to need to execute, so your production organization ought to stay the same. You're still going to need to troubleshoot the process, so your Manufacturing Sciences process support stays the same. You still need scheduling, you still need downstream recovery and purification. All that stays the same.

I Still Have A Few Questions...

  • Do Single-Use Bioreactors (SUBs) decrease my plant shutdown times?
  • What is the turn-around time (from dirty to prepped) on a SUB?
  • What commercial products are manufactured with SUBs?
  • At what scale are SUBs proven?
  • What is the Total Cost of Ownership of SUBs?
If you want to know more, Dan over at EMD Millipore has some answers.

Contact Dan For More Info

Thursday, August 23, 2012

Biologics Manufacturing - Great Amgen Video

Here's a really informative video on biologics manufacturing.



Or if you prefer to watch this on YouTube, start at 15 seconds in and you can stop watching at 3:28 where the talking points begin.

It's the same old talking points that I've addressed:
My questions for innovator drug manufacturers are as follows:
  1. Is your Master Cell Bank single-cell sourced?
  2. You have never upgraded your cell line (v2.0) for an existing product, correct?
  3. Are you willing to publish the CofA testing requirements of your clinical material and abide a regulatory body approving/rejecting drugs according to whether or not each batch meets/fails those CofA specs?
Your YES response to all these questions will stand me corrected.

All the propaganda aside, nice video.

For more information on biologics manufacturing, see Amgen's biotech learning resource.

Wednesday, August 1, 2012

Follow-on Biologics Technology as of 2008

A lot of people wonder why I blog about biosimilars.  I don't have a Ph.D and I actually don't have any vested financial interest in the biosimilars.

Biosimilars have entered the mainstream ever since their mandate was specified in the Patient Protection and Affordable Care Act (a.k.a ObamaCare).

Of the many things that ObamaCare mandated, one of them was that the FDA provide a regulatory approval pathway for biosimilars.  Imagine that... a mandate for government.

In the United States as of the publication of this post, there is no way to introduce a biologic onto the market unless you are the person who came up with it... i.e the "innovator."

That's right.  Pharmaceutical companies research the the drug, show safety/efficacy in the clinic, demonstrate cGMP compliance get the FDA nod to market the drug in the United States.

But when pharma drug patent(s) expire, other pharmaceutical manufacturers are permitted to seek FDA approval to sell the generic version... so long as they meet the rigors of FDA facility inspection and their application shows that they can produce the active pharmaceutical ingredient (API).

How do they know how to make the drug?  The process isn't that simple, but you can figure it out from the aforementioned drug patents.

So why can't this be done for biologics?

The reason this putatively can't be done for biologics is because the active pharmaceutical ingredient is not made by a chemical reactions managed by humans.  The API is made by chemical reactions managed by genetically engineered cells.
  • For small biologics like insulin or hGH, E.Coli or other bacteria is capable of producing.  
  • Large biologics require mammalian cells like Chinese Hamster Ovary (CHO).
So when the FDA approves of the manufacturing procedure for a biologics process, the approval goes for that specific genetically engineered cell line.  Not even the innovator can switch out the cell line without going back to the clinic.  Them's the rules.

It stands to reason the cell line is locked for the innovator, it is locked for everyone else, hence a legal monopoly for biologics in the United States.

But back to the reason I blog about biosimilars.  In 2008, I ran into an old friend (we'll call Morpheus) who worked at a big U.S. biotech company and he asked me if I remembered a mutual friend (who we'll call Neo).  Back in the early 2000s, Neo had left to work for an overseas pharmaceutical company to lead their biologics development.

Morpheus was telling me that Neo's company had produced an exact copy of one of his company's flagship drug.

Me: "What do you mean, exact?"

Morpheus: "I mean it's closer to our clinical controls than our current manufacturing process."

What Morpheus was saying was that the competitor's drug matched the drug used to produce the successful clinical trial better than what the U.S. biotech company was manufacturing in 2008.

What was on everyone's minds at the time was industrial espionage.  After all, it was against prevailing dogma that anyone could reproduce the biologic.  But to replicate the drug better than the innovator... is insane.

I never heard about the allegations of industrial espionage again.

But what I do know is that the term biosimilars is a misnomer.  They ought to be called, "Bioidenticals."

And this was back in 2008.  Who knows how far these overseas companies pushed this technology?

One thing for certain is that the technical hurdles of protecting the biologics monopoly are falling.  And with the ObamaCare mandate for a regulatory pathway for biologics, it appears that the regulatory hurdles are falling as well.

See Also:

Tuesday, July 24, 2012

More Propaganda on Biologics

From Forbes' article on biosimilars:

Compounding the complexity, even biologics that appear chemically the same have unique structural fingerprints as a result of the way protein structure assumes its functional shape, a process known as “folding.” Because biosimilars are so difficult to develop, they sometimes don’t work as effectively as the innovator biologic drug.

I wonder if the author knows how innovator biologic drugs are manufactured.  If he did, he'd know that these drugs are produced in batches and that controls must be in place to ensure that the drug product is the "same" from batch-to-batch.

Biologics are complex due to their size.  Their efficacy depends on not just their chemical composition, but on their shape.  But there are ways to provide a high degree of assurance that the drug product is effective.

In computer science, there's a concept called, "Checksum."  A checksum is a "fingerprint" for digital data... no two pieces of digital data produces the same "fingerprint."  In the world of QC drug testing, there is a concept called peptide mapping.  This is where you digest the protein with enzymes, chopping it at specific places.  Then you take the chopped-protein and run it through a chromatography column and see what order the parts come out.  The order the parts come out is a "fingerprint" for the molecule.

As for the activity of the drug product, you can test that a biosimilar trastuzumab binds to the HER2 protein.  I mean, that's how they tell if you are HER2+ in the first place.

I'm not saying that biosimilars ought to get rubberstamped with lower standards.  I'm saying that the vast majority of biologics manufacturing requirements of the innovator drugmaker can be ported over to biosimilars and that these putative hurdles described by our journalists are dated and not applicable given the modern technology available.

See also:

Monday, May 21, 2012

Is @SAFEbiologics Really About Safe Biologics?


In a prepared statements made to the FDA on biosimilars, the Alliance for Safe Biologic Medicines (@SAFEbiologics) outlined 5 areas for deeming biosimilars to be interchangeable with the original drug [PDF]:

  1. Clinical testing
  2. Global supply chain and manufacturing monitoring
  3. Track, trace and naming
  4. Clear labeling and packaging
  5. "Close deliberate scrutiny"

There aren't many involved in the current system of drug approval that will disagree with these five points. These steps are known to add cost to biosimilar manufacturers thereby making the biosimilar less competitive.

cGMP regulations of finished pharmaceuticals already cover manufacturing monitoring. As well, 21 CFR Part 211 Subpart G issues six sections of regulations that cover labeling and packaging. You can be certain that the other 3 items are also required of existing cGMP manufacturers.

What the Alliance for Safe Biologic Medicines appears to be urging the FDA is simply this:

Regulate biosimilars the same way that you regulate original biologics.
Which isn't a surprise; nor is this request unfair.

What is a surprise is how ASBM's chairman, practicing endocrinologist Dr. Dolinar, doesn't understand the Central Dogma of Molecular Biology:

Biologics are complex, large molecule drugs that are grown inside living cells using unique and proprietary processes. For this reason, no two biologics made from different cell lines or using different processes can be identical based on today's science.

This is simply not true in most cases. We know that the DNA is DNA and that cellular machinery for any biological organism can transcribe and translate that DNA into protein.

Two insulin molecules made from different cell lines (one E.Coli, the other human cells) or using different processes are, in fact, identical based on today's science.

In fact, the opposite of Dr. Dolinar's statement is exactly how we have a thriving biotech industry in the first place: that biologics made by microbes in big stainless steel bioreactors is equivalent to the large complex biological molecules produced by human cells in vivo.

Here's another doozy:

Biologics are also highly sensitive to the manufacturing process. In fact, altering a single manufacturing parameter can change a compound's identity and/or the precise effect it has on the human body.

This statement is pretty bizarre as well. Here's why:

Original biologics manufacturers are constantly changing their manufacturing processes... or worse, their manufacturing processes changes on them. At this very moment, FDA-approved cell culture processes are being upgraded... with changes to media and operating conditions. Some manufacturers are even changing the cell line and hoping that they don't have to go back to the clinic.

Secondly, manufacturing processes where altering a single manufacturing parameter can change product quality is not a viable process at all. I don't dispute that these processes existed; I dispute that they are passing regulatory muster and being approved by the FDA today.

The robustness of a process can be defined as how many changes the manufacturing process can endure before the product quality or productivity suffers. It's the year 2012 and back in 2007, process scientists were working on process capability and trying to understand parameter interactions such that this proverbial single change in a manufacturing parameter cannot alter the the identity of the product.

Lastly, the FDA has been pushing programs like Process Analytical Technologies (PAT) and Quality by Design (QbD) to explicitly avoid situations where changing a single parameter renders the process incapable of meeting product quality specs. Most of my customers have been applying these principles for years.

ASBM's chairman has talking points from the late 90's: his information was true at one point in time but has since become hyperbole.

Why is there an ASBM in the first place and why is their chairman publicly issuing misleading hyperbole? Let's have a look at ASBM's website (safebiologics.org)...and there you have it:

ASBM members
Genentech... Amgen... Biotechnology Industry Organization (Big Biotech's lobby).

I'm pretty certain that these guys truly have the good intentions of protecting patient safety.

I'm also pretty certain that these guys truly have the intentions of protecting their status-quo legal monopoly by throwing regulatory hurdles in front of their future competitors.

Further Reading


FDA Releases Draft Guidance on Biosimilars


Tuesday, March 27, 2012

Cornell ChemE Prof Makes Glycoproteins Without CHO Cell Culture

Just out yesterday:

Cornell Chemical Engineering Professor Produces Glycoproteins with E.Colicornell article

The thrust of the story is that a Cornell professor, Matthew DeLisa, has figured out how to make glycoproteins using E.Coli. Readers of my blog know that the reason we humans use Chinese Hamster Ovary (CHO) cells to make human glycoproteins is because only mammalian cells can do post-translational glycosylation while the simpler bacteria cannot.

This is where Dr. DeLisa says, "Not anymore."

You'll have to read the paper from his Nature paper (included at the bottom) if you want to get wonkish, but described in lay terms, the Cornell research team added:


  • Four enzymes from yeast cells:

    1. Yeast uridine diphosphate-N-acetylglucosamine transferases Alg13
    2. and Alg14
    3. Mannosyltransferases Alg1
    4. and Alg2
  • One enzyme bacterial from from Campylobacter jejuni
    oligosaccharyltransferase PglB

to E.coli cultures to get the desired glycan structures. All this, I presume, involves more than cleverness and advanced pipetting skills.

Sounds quite promising as the tech is being commercialized through a startup called Glycobia. As science is skeptical, so is this fermentation engineer (of the commercial value of this venture).

The process economics of CHO vs. E.Coli does not clearly point in the direction of bacterial cultures. If you look at two nearly identical drugs, Lucentis (made with E.coli) vs. Avastin (made with CHO), you have drastically higher cost for the E.coli product. Lucentis is the Fab region of the antibody while Avastin is the whole antibody (Fab + Fc) and the costs are $2,000 vs $150 (assuming the same concentration of API gets the job done). The markup has little to do with Genentech being greedy.

A quick Google search of "cho vs e coli process economics" will get you to a book by Ajit Sadana (1998) on Bioseparation of proteins. Starting on page 66, he goes through an example of Activase (tPA) made in CHO vs. E.coli.

CHO had 5 steps while E.coli had 16. CHO had a 47% yield while E.coli had a 2.8% yield... primarily due to the extra recovery steps to remove the endotoxin that E.coli creates that CHO does not. Sure, this example is 1998 technology talking about (likely small scale) purification, but I have yet to see the process economics work in favor of E.coli for biologics.

If contamination concerns (like the Genzyme Allston plant cited) are the main cost avoidance, I'm going to come out and say that this research will remain academic: bioreactor contaminations are easy to prevent when management is committed to contamination reduction.

If replacing the known quantity that is CHO with an unknown quantity that is E.coli + Bottom-Up Glycoengineering (BUG) technology is all we get (i.e. without orders of magnitude increase in culture titers or reduction in variability), then my money with CHO.

More reading:


Thursday, February 16, 2012

FDA Releases Draft Guidance on Biosimilars

A week ago, the FDA released three documents to comply with the Biologics Price Competition and Innovation Act (BPCI Act) to create competition within the biologics space.

  1. Q&A for BPCI Act
  2. Scientific Considerations of Biosimilarity
  3. Quality Considerations of Biosimilarity
Why is the bankrupt U.S. government passing more laws to empower the FDA on this matter? Basically because the markets created biologics faster than the FDA was able to respond and this is essentially catch-up.

You see, unlike small biological molecules, large biological molecules (called "biologics") cannot be feasibly or economically synthesized with chemical reactions. Biotech companies differ from pharma companies because they genetically engineer microbes to secrete the complex biological molecules and then produce a lot of it with fermentation or cell culture.

The FDA has long held:

Quality cannot be tested into the product

I was at this IBC Conference once where a professor got up and said, "No one knows this to be more true than academia... each year we test our students more and more, and each year they don't get any smarter."

It has long been insufficient for drug companies to produce a drug that meets product quality specifications when substitute raw materials were used or procedures not followed during the manufacture of the drug.

Applying this rule to biologics, it would hold that anyone who doesn't have the original cell line used to manufacture the name-brand biologic would violate this FDA dogma and thus be forbidden from selling their biologic in the US markets.

In essence, there's no way for the FDA to allow biosimilars into the US markets without throwing everything they've been doing out the window. This lack of a regulatory pathway forbids biologics made by companies other than the original manufacturer to be sold in the U.S, thereby handing US biotechs monopolistic power.

This is why there's a BPCI Act in the first place. It is to force the FDA to create a pathway for generic biologics to enter the US markets and induce competition.

Watching the FDA on biosimilars has become a spectator sport for the wonkish Regulator Affairs folk. The folks over at Bioprocess Blog cover this much more thoroughly.

Suggested reading:


-->