PETRI DISH PERSPECTIVES: BIOTECH UNLEASHED

Episode 11: Genentech

• Manead Khin • Season 1 • Episode 11

Send us a text

🧬 From VC to Blockbusters: The Genentech Story
In Episode 11 of Petri Dish Perspectives: Biotech Unleashed, host Manead dives into the legendary rise of Genentech, the startup that sparked the biotech revolution. From its humble beginnings in a borrowed lab to pioneering insulin, tPA, Herceptin, and more, learn how Genentech turned recombinant DNA into real-world cures. Discover the bold VC bets, game-changing antibodies, and the Roche acquisition that preserved its science-first culture. A must-listen for biotech nerds and curious minds alike!

🎧 #Genentech #BiotechPodcast #Herceptin #Avastin #Rituxan #RecombinantDNA #OncologyInnovation #PetriDishPerspectives #HealthcareInnovation #PharmaHistory #RocheGroup #StartupToSuperpower

Support the show

© 2025 Petri Dish Perspectives LLC. All rights reserved.

🎙️ Intro

Hello and welcome to Petri Dish Perspectives: Biotech Unleashed, the podcast where we geek out about science and the companies shaping the future of healthcare. I’m your host, Manead, and I’m a scientist with a PhD background in cancer biology and analytical chemistry. With every episode, my goal is to deliver digestible pieces of information on healthcare companies under 30 mins. Also a quick sidenote, I cannot believe we’re on episode 11. Thank you for all the support so far and I’m so grateful for it!

Today’s episode is about one of the most iconic names in biotech history, Genentech. This is also a follow up from the Roche episode since Genentech is one of the companies that Roche acquired. Founded in a lab over a beer and a big idea, Genentech didn’t just help create an industry, it became the poster child for what biotech could do. From synthetic insulin to cutting-edge cancer antibodies, Genentech revolutionized how we make and deliver medicines. I had a ton of fun researching this episode so I hope all of you can enjoy it!

Quick disclaimer: full credit goes to all original sources cited in the transcript.

Grab your coffee or tea, settle in, and let’s jump right in.

🧬 Segment 1: The Birth of Biotech – Genentech’s Founding Story

Let’s rewind to 1976, in the heart of Silicon Valley, a place more known at the time for microchips than monoclonal antibodies. This is where Genentech was born, not in a polished pharma boardroom, but from the bold vision of two very different minds.

One of them was Robert A. Swanson, better known as Bob Swanson, a man who had, dare I say, the audacity to bet on biology when few in business thought it was worth the risk. His story is especially inspiring to me, since I’ve also had a run in the biotech VC world and his journey is the kind of founder story that gets me fired up.

Swanson wasn’t a scientist. He was a young venture capitalist, barely in his late 20s, working at Kleiner Perkins, the now-legendary Silicon Valley firm. It was the mid-1970s before the words “biotech” or “startup” really meant anything. But Swanson had been digging into recombinant DNA technology and was convinced it could become something big. Recombinant DNA is a form of DNA created in a laboratory by combining genetic material from two or more different sources. This process, often referred to as genetic engineering or biotechnology, involves isolating and manipulating DNA sequences and then inserting them into a vector (like a plasmid) to be replicated in a host organism.

At the time, the buzz around genetic engineering was still confined to university labs and niche conferences. Swanson saw commercial potential where others didn’t. He pitched the idea to his partners at Kleiner Perkins but they weren’t interested. In fact, they let him go. But that rejection turned into redirection.

Later that year, Swanson reached out to Herb Boyer, a biochemist at UCSF who had co-discovered a method for splicing genes into bacteria, essentially teaching microbes how to produce human proteins in their own systems. Swanson cold-called him and asked for just ten minutes of his time.

That ten-minute coffee meeting turned into three hours, and by the end of it, Genentech was born. The name Genentech is a portmanteau, derived from the words "genetic engineering technology". It was created by one of the company's founders, Dr. Herbert Boyer, as a way to reflect the company's focus on utilizing genetic engineering techniques. 

Swanson brought the business instinct, the drive, and the fundraising grit. He pulled together legal and regulatory frameworks that didn’t yet exist for biotech, and famously helped broker the first deal with Eli Lilly to produce human insulin using recombinant DNA, which would go on to become the first-ever biotech drug.

But Genentech’s first proof-of-concept wasn’t insulin. It was somatostatin, a human growth hormone regulator. They successfully cloned and expressed the gene in E. coli, and in 1977, published the results. The world was stunned that this tiny upstart had actually programmed bacteria to make a human protein.

Then came the blockbuster: insulin. In 1978, Genentech announced it had synthesized recombinant human insulin, a breakthrough that would revolutionize diabetes care. They licensed the product to Eli Lilly, which brought Humulin to market in 1982. It was the first recombinant biotech drug ever approved by the FDA. Also side note, if you want to hear more about insulin and commercialization, I highly recommend checking out Episode 2 on Eli Lilly.

Swanson served as Genentech’s first CEO, leading the company through technical challenges, public skepticism, and eventually to its IPO in 1980, which raised $35 million in just minutes and ignited the first-ever biotech IPO boom.

But Genentech was more than a company. It was a cultural shift. It combined the urgency of a startup with the rigor of academia. It had an open-lab environment, flat hierarchies, and strong scientific authorship. Scientists weren’t just employees, they were co-creators.

Swanson’s legacy is that of a bridge-builder, connecting the capital of Silicon Valley with the science of molecular biology. Without him, Genentech might have remained just another academic project. Instead, it became the blueprint for modern biotech.

Sadly, Bob passed away in 1999 at just 52, after battling a brain tumor. But his vision that ideas in the lab could change the world, with the right science and the right business model lives on in every biotech company that followed.

🧪 Segment 2: Pioneering Proteins – From Hormones to Blockbusters

Let’s rewind to the late 1970s and 1980s, a time when biotechnology wasn’t even a proper industry yet, but Genentech was laying down the foundation, one molecule at a time.


📏 Protropin (Somatrem) – Synthetic Human Growth Hormone

Fresh off the insulin breakthrough, Genentech’s next mission was tackling growth hormone deficiency in children. Until then, the only way to get human growth hormone (hGH) was to extract it from human cadaver pituitary glands, a process that was limited in supply and eventually linked to the spread of Creutzfeldt–Jakob disease (a rare and fatal brain disorder).

Using similar recombinant DNA methods, Genentech cloned the hGH gene and inserted it into bacteria. The result: Protropin, approved in 1985, a synthetic version of hGH, identical in function to the natural hormone.

This was not only a safer alternative but also infinitely more scalable. Protropin gave children with growth hormone deficiency access to a consistent, reliable therapy and no cadaver glands required. It was Genentech’s first FDA-approved product under its own name and marked its evolution from a science lab into a commercial biotech force.


❤️ Activase (tPA) – Clot-Busting in Real Time

While the first two products addressed chronic endocrine conditions, Genentech’s next big move came in acute care, specifically, cardiovascular emergencies.

Scientists had long known that tissue plasminogen activator (tPA) could dissolve blood clots by activating the body’s own clot-busting system. But sourcing enough tPA from human tissue was impossible. So Genentech again turned to recombinant DNA technology, cloning the tPA gene and producing the protein in Chinese hamster ovary (CHO) cells.

In 1987, Activase (recombinant tPA) was approved to treat acute myocardial infarction (heart attacks). It was later expanded to pulmonary embolism and ischemic stroke.

What made Activase groundbreaking wasn’t just the science, it was how it redefined emergency medicine. Paramedics could administer it in ambulances to stop heart attacks in progress. ER doctors could use it to save stroke victims. It was one of the first biotech products used in critical care settings, not just for long-term disease management.

Although controversial due to its high cost and debate over effectiveness in stroke, Activase helped cement Genentech as a player not just in biology, but in life-or-death, frontline medicine.

These early successes weren’t just product wins, they were proof-of-concept moments for the entire biotechnology field. Genentech demonstrated:

  • Recombinant DNA could safely replace animal/human-derived therapies
  • Biotech products could scale commercially and hit blockbuster revenues
  • Targeted biologics could change how we approached immune and chronic diseases

By the late 1990s, Genentech had earned its reputation as the first true biotech company, a company that not only discovered new molecules but built the infrastructure, regulatory precedent, and manufacturing systems to make biotech a real industry.

💊 Segment 3: Oncology Revolution – Herceptin, Avastin, and Beyond

By the early 2000s, Genentech wasn’t just riding the biotech wave, they were shaping it, particularly in oncology. Their next generation of therapies would redefine what targeted cancer treatment looked like.

Let’s begin with Herceptin (trastuzumab), approved by the FDA in 1998. This wasn’t just another cancer drug, it marked a turning point in oncology. Herceptin targets HER2, a receptor protein that is overexpressed in about 20 to 25 percent of breast cancers. Before Herceptin, HER2-positive breast cancer was one of the most aggressive and deadly subtypes with limited treatment options.

The story behind Herceptin started at UCLA in the early 1980s. Researchers Dennis Slamon and Axel Ullrich discovered that amplification of the HER2 gene led to overexpression of its protein product in a subset of breast tumors. This insight turned into a hypothesis: could a monoclonal antibody be used to block HER2 and stop tumor growth?

Genentech took this idea and ran with it. After years of development and trials, they produced trastuzumab, an antibody that could bind to HER2 and inhibit signaling pathways that fuel cancer cell proliferation. Clinical trials showed that when combined with chemotherapy, Herceptin dramatically improved outcomes for HER2-positive patients.

But that was only part of the innovation. Roche Diagnostics also developed a companion diagnostic test to identify HER2-positive tumors. This was one of the first instances where treatment and diagnosis were developed in tandem, launching the era of personalized cancer therapy.

Next is Avastin (bevacizumab), approved in 2004 for metastatic colorectal cancer. Its mechanism of action was groundbreaking. Rather than targeting the cancer cells directly, Avastin blocks VEGF, a protein that tumors release to grow new blood vessels. By cutting off their blood supply, tumors essentially starve and stop growing.

The origins of Avastin trace back to Napoleone Ferrara, a researcher at Genentech, who isolated VEGF in the late 1980s and proved its role in angiogenesis, the formation of new blood vessels. This discovery led to the hypothesis that stopping VEGF might inhibit tumor growth. Ferrara and his team developed bevacizumab, a humanized antibody that neutralized VEGF, and after promising preclinical data, it moved into trials.

Avastin became the first anti-angiogenic therapy ever approved. Over time, it gained approvals in several other cancers, including lung, kidney, brain, and cervical. Though it faced some regulatory pushback, especially around its use in breast cancer, Avastin proved that starving a tumor’s lifeline was a valid therapeutic strategy.

And then there’s Rituxan (rituximab), which became the first monoclonal antibody approved to treat cancer in 1997. Rituxan targets CD20, a protein found on the surface of B cells. It was developed to treat non-Hodgkin lymphoma by tagging malignant B cells for destruction by the immune system.

The discovery began with a small biotech startup called IDEC Pharmaceuticals. In the early 1990s, IDEC scientists, in collaboration with researchers at Stanford, engineered a chimeric antibody that combined mouse variable regions with human constant regions to reduce immune rejection. This novel design allowed rituximab to be safely used in humans.

When Genentech partnered with IDEC to co-develop and commercialize Rituxan, it quickly demonstrated remarkable efficacy in lymphoma and became a standard of care. But Rituxan’s impact extended beyond oncology. It was later approved for autoimmune conditions like rheumatoid arthritis, granulomatosis with polyangiitis, and other diseases driven by overactive B cells.

Its success proved monoclonal antibodies could work across therapeutic areas and inspired an entire generation of antibody-based therapies, including successors like Gazyva and biosimilars.

Together, these therapies, Herceptin, Avastin, and Rituxan, became known as the “big three.” By the early 2010s, they were pulling in over $20 billion annually and accounted for the majority of Genentech and Roche’s oncology revenue.

These weren’t just financial hits, they shaped clinical practice guidelines, trained a generation of oncologists in targeted therapies, and laid the groundwork for modern immuno-oncology.

🧬 Segment 4: The Roche Acquisition – Biotech Meets Big Pharma

All this success caught the attention of Roche, which had first invested in Genentech in 1990, acquiring a controlling stake. But in 2009, they went all in, purchasing the remaining shares for $47 billion. It remains one of the largest biotech deals in history.

But what made the Roche–Genentech deal unique was how Roche handled the integration.

Rather than folding Genentech into a traditional pharma structure, Roche let Genentech maintain its scientific independence. The South San Francisco campus remained the hub for early-stage research, while Roche provided global commercialization and regulatory support.

This “dual operating model” worked. Genentech’s startup-style culture continued to attract top-tier scientists, while Roche scaled manufacturing, trials, and international launches.

Together, they began launching a new generation of biologics: Perjeta, Kadcyla, Alecensa, Tecentriq, and many of them building directly off Genentech’s earlier work.

Roche’s diagnostics division played a key role here too. Their ability to develop biomarker tests in parallel with Genentech’s biologics meant that new drugs came to market with companion diagnostics, ensuring they reached the right patients faster.

This synergy of biotech innovation + pharma muscle + diagnostic precision, became the gold standard. Few companies could replicate it.

By the mid-2010s, Roche and Genentech were consistently ranked among the world’s most innovative healthcare companies.

👩‍🔬 Segment 5: People Who Made Their Mark

Genentech’s culture was deeply shaped by the personalities who led it. Aside from the founders, I’d like to highlight Art Levinson.

By the mid-1990s, Genentech had already helped birth the biotech industry with recombinant insulin and growth hormone. But the next chapter would demand a different kind of leadership, someone who could scale the company into a pharmaceutical powerhouse without losing its scientific edge or startup spirit.

Enter Arthur D. Levinson, better known as Art Levinson.

A molecular biologist by training with a PhD from Princeton, Levinson joined Genentech in 1980 as a postdoctoral researcher working under pioneering scientists like Herb Boyer. He rose quickly through the ranks, not because of boardroom bravado, but because of his deep belief in the science and the people doing it.

When Levinson became CEO in 1995, it marked a rare moment in biotech: a bench scientist taking the helm of a multibillion-dollar company. But Levinson wasn’t your typical executive. He knew how research worked, he knew the power of culture, and he had the strategic clarity to guide Genentech through its most ambitious era yet.

One of the most pivotal challenges of Levinson’s tenure came with Roche’s increasing stake in Genentech. Roche had been a majority shareholder since 1990 but had allowed Genentech to operate independently. By the mid-2000s, Roche wanted full ownership and in 2009, it launched a $47 billion buyout.

Levinson fought hard to preserve Genentech’s scientific culture during these negotiations. He was adamant that the company’s research independence and innovation-first mindset must be protected.

Thanks to his leadership, Genentech secured an unusual deal: despite becoming a wholly owned subsidiary of Roche, it would retain significant autonomy, especially in R&D. Its South San Francisco campus remained the HQ for global biotech operations, and many of its scientific leaders stayed on to continue shaping its pipeline.

After stepping down as CEO in 2009, Levinson didn’t disappear. He served as Chairman of Genentech and later joined Apple’s board of directors, where he became Chairman in 2011, succeeding Steve Jobs. That tells you just how respected he was across both science and tech.

Levinson's leadership style, that was quiet, principled, scientifically driven, stands in contrast to the flashier narratives of Silicon Valley. But his impact is undeniable. He built one of the most respected R&D cultures in the world, proved that a scientist could lead at scale, and ensured Genentech’s identity survived even after a historic acquisition.

Today, Genentech continues to be led by scientists, many with MDs or PhDs, a reflection of its belief that science should lead strategy.

🧭 Segment 6: What’s Next for Genentech?

Genentech’s current pipeline reflects the next frontier of medicine.

  • Immuno-oncology remains a core focus, with drugs like Tecentriq (an anti–PD-L1 checkpoint inhibitor) being tested in new combinations and tumor types.
  • Cell and gene therapy is gaining momentum. Roche’s acquisitions of Spark Therapeutics and Tusk Therapeutics bring AAV gene delivery and Treg immunotherapy into the mix.
  • Neurology is a growing focus, with Ocrevus for MS, Enspryng for NMOSD, and multiple Alzheimer’s and Parkinson’s programs underway.
  • Genentech is also advancing RNA-based therapies, bispecific antibodies, and targeted protein degraders, exploring new ways to drug the undruggable.

More recently in May of 2025, Genentech has announced a $700 million investment to build a high-volume fill/finish manufacturing facility in Holly Springs, North Carolina, which is the first in the state. The project will create 420 jobs and generate an estimated $1.2 billion economic impact, supported by Holly Springs’ strategic growth as a life sciences hub. 

As of June 2025, Genentech doesn’t trade separately from Roche, but the Roche Group maintains it as a fully-owned subsidiary, with its U.S. market presence and brand identity fully intact.

Of course, since Genentech is a subsidiary of Roche, it doesn’t have its own stock. However, Roche’s stock sits at $40.91 a piece, with a total market cap of $266.84 billion and honestly much of that value rooted in the innovation coming out of Genentech. Genentech has over 13,000 employees and has multiple locations, with its primary location in South San Francisco, CA. The company has a large U.S. footprint, including 13 manufacturing and 15 R&D sites. According to Glassdoor, an average salary for a PhD at Genentech varies depending on experience and specific role, but generally ranges from $112,000 to $300,000 per year. 

🧩 Segment 7: Keys to Genentech’s Success

So what makes Genentech such a biotech legend?

  • First-mover advantage: From recombinant insulin to antibodies, they were always first.
  • Science-first culture: Leadership by scientists, not just MBAs.
  • Academic collaboration: Deep ties to UCSF, Stanford, and research institutions.
  • Startup roots: Even as they scaled, they kept the fast-paced, mission-driven energy.
  • Strategic independence: Even post-acquisition, Genentech remains creatively autonomous. But part of me does wonder whether Genentech would’ve been successful on its own or Roche was necessary for the increasing success. We will never know…

Regardless, together with Roche, Genentech created the blueprint for modern biotech and they’re still evolving it every year.

🎵 Outro

🎵 [Uplifting music begins]

That’s a wrap on today’s Petri Dish Perspectives! I hope you enjoyed this deep dive into Genentech from its startup beginnings in a borrowed lab to its status today as a cornerstone of cancer therapy and biotech innovation.

If this story inspired you, please subscribe, leave a review, and share with your fellow biotech nerds. Got a company you want covered next? Please let me know I’d love to do the research and report back to y’all.

Until next time, stay curious and keep exploring the science reshaping our world.

🎵 [Music crescendos and fades out]


References


  1. www.wikipedia.org
  2. www.genentech.com
  3. https://www.kleinerperkins.com/case-study/genentech/ 
  4. www.finance.yahoo.com 
  5. www.glassdoor.com 
  6. https://www.hollyspringsnc.gov/CivicAlerts.aspx?AID=1607 

People on this episode