Tag Archives: books

RESI Boston Webinar Series: Your Roadmap to a Stronger Pitch, Smarter Partnering, and Greater Investor Engagement 

8 Jul

By Max Braht, Director of Business Development, LSN

Max-Braht-HeadshotRESI Boston returns September 17 to 19, 2025, and in the lead-up to the event, Life Science Nation is offering a series of free educational webinars designed to help life science startups strengthen their pitch, navigate partnering, and engage more effectively with investors. Whether you are pitching at the Innovator’s Pitch Challenge, booking one-on-one meetings, or attending RESI for the first time, these sessions offer valuable tools to get the most out of your conference experience.

July 10, 2025 at 12:00 PM
Standing Out to Investors: How to Tell Your Startup Story for Maximum Impact – Sign Up
The most successful entrepreneurs are powerful storytellers. This interactive bootcamp will show you how to shape and share your startup’s story for maximum investor impact. Learn how to sharpen your elevator pitch, refine your 12-slide deck, and make your messaging resonate with different stakeholders in the deal chain. Participants will leave with actionable language tools and techniques to boost their confidence and communication skills.


July 22, 2025 at 12:00 PM
Tips on Pitching: From the Application to the Q and A – Sign Up
Get a firsthand look at what makes a pitch resonate with investors. Join RESI judge, Bruce Cohen, who has sat on both sides of the table as an investor and an Innovator’s Pitch Challenge participant, for an inside perspective on how to stand out. This session covers best practices for your application, how to deliver a clear and convincing pitch, and how to prepare for and respond to investor Q and A.


August 7, 2025 at 12:00 PM
Biotech Exec Webinar – Who should be on your executive team? Structuring for success and fractional execs. – Sign Up
Join Life Science Nation (LSN) and Biotech Exec in early August for a free webinar on how to structure your life science startup executive team for success. Startups can fall victim to many strategic mistakes even when the science holds up. Choosing the right indication. Performing the right assay. Designing the right trial. Structuring a fund raise optimally. But one of the least talked about mistakes is putting the wrong people on the executive team or simply not brining on the right talent. We will have an engaging discussion that will include case studies of how things went wrong and testimonials from experienced CEOs and investors regarding what went right and what went wrong in their previous experience. Investors have a knack for knowing immediately if the executive team is poised to go the distance. Learn how to avoid the pitfalls most investors look for to ensure your barrier to fundraising is at a minimal. Learn if a fractional exec is right for you or if you should be looking for full-time right away.


August 18, 2025 at 12:00 PM
Partnering Tutorial: Making the Most of the RESI System – Sign Up
RESI’s partnering system is a powerful tool—but only if you know how to use it. Join the LSN team as we walk through how to navigate the platform, target the right investor fit, manage your outreach, and make the most of every meeting. We will also share our recommended strategy for follow-ups and how to leverage RESI content to support your fundraising journey.


September 4, 2025 at 12:00 PM
Investor Fireside Chat: Fundraising in Today’s Biotech Climate – Sign Up
Hear directly from investors about what is driving deal flow in the life science ecosystem. This fireside chat will cover current investment trends, what investors are looking for in early-stage biotech companies, and how startups can better position themselves to raise capital. Bring your questions and gain insight from experts who know what makes a company fundable.

Registration is open for all webinars. These sessions are free to attend and are designed to help you prepare for meaningful investor interactions and set yourself up for success at RESI Boston this September.

Announcing Premier PLUS for RESI Partnering  

1 Jul

By Max Braht, Director of Business Development, LSN

Max-Braht-HeadshotPre-pandemic I came across a survey that went out to in-licensors and investors to see from where they sourced their deals. Pipeline databases? Literature? Partnering meetings? Medical conferences? Incoming email solicitations? Website submissions? Warm intros? As it happens, the #1 answer was partnering events, representing the first point of contact for the two parties that eventually made a deal. Even the BioNTech and Pfizer deal came out of a partnering conference in 2013.

However, partnering conferences require a lot of targeted effort in the partnering system to reach out to potential partners and follow-up on the initial outreach. For many early-stage life science entrepreneurs, preparing for a RESI Conference means juggling multiple priorities — investor research, updating pitch decks, team coordination, and most importantly, the execution of your investor outreach strategy.

Many entrepreneurs underestimate the time and effort it takes to identify the right targets, write effective messages, and follow up with these targets consistently. Without a strategic approach, it’s easy to miss out on valuable meetings.

To address this, LSN is introducing a new service: Premier PLUS Partnering, where we do the partnering for you.

Premier PLUS Partnering provides hands-on support from the LSN team to manage your partnering outreach from start to finish.

Drawing on years of experience helping thousands of companies connect with investors, LSN can assist by:

  • Identifying and reaching out to relevant investors and partners
  • Crafting consistent, professional messages tailored to each contact
  • Managing communication and scheduling through the partnering platform
  • Ensuring confirmed meetings in advance of the conference

For entrepreneurs who know the importance of effective partnering strategies and are interested in taking advantage of Premier PLUS Partnering at RESI Boston September, please reach out directly to the LSN team at sales@lifesciencenation.com or include the add-on in your RESI Boston September Registration.

Pullan’s Pieces #1 – Organ on a Chip

1 Jul

Acceleration of laboratory-based technical and computational cross-fertilization, and ethical and cost pressures on regulatory bodies and therapeutic innovators is driving advancements in preclinical human-based technologies.

Organ (Lab)-on-chip (OoC/LoC) is one of the most striking examples of new translational research technology expansion with ~35% CAGR expected over the next decade (below).  

Collaborations between academia and CRO’s are driving improvements in organoid technology for the field of oncology broadly and are expected to improve OoC adoption.  Academic innovation using commercial OoC technology is also advancing applications in specific indications in oncology.  CRO’s continue to build off established uses in ADME and toxicology to explore R&D applications in oncology space and have even combined organ systems to support elaboration of multiple drug parameters in a single assay.

DEALS

The Tara Biosystems – Valo Health deal is a nice example of how an organ-on-a-chip technology approach has driven collaborations, acquisitions and deals:

  • Tara Biosystems and GSK collaborate on CV drug profiling (2019)
  • Valo Health acquires Tara Biosystems for CV OoC platform (2022, ~$75M upfront)
  • Valo and Novo Nordisk sign CV drug discovery deal (2023, $60M upfront, $2.7B total)

EmulateTissUse and Mimetas have also been backed by strong big pharma collaborations (AstraZeneca, Bayer, Roche) and funding rounds.

The Needle Issue #9

1 Jul
Juan-Carlos-Lopez
Juan Carlos Lopez
Andy-Marshall
Andy Marshall

Drug development efforts targeting the constitutive 26S proteosome have led to the development of several important multiple myeloma (MM) and mantle cell lymphoma treatments, including the first landmark FDA approval of Millennium Pharmaceuticals’ (now Takeda) dipeptide boric acid Velcade (bortezomib) in 2003 and second-generation molecules, such as Amgen/Ono Pharmaceutical’s irreversible inhibitor Kyprolis (carfilzomib) and Takeda’s orally available inhibitor Ninlaro (ixazomib). Second-generation versions of these ‘pan-proteosome’ drugs have longer duration of effect, reduced peripheral neuropathy and increased safety in renally impaired patients, but may cause gastrointestinal and cardiac toxicity. This toxicological profile has shifted attention to developing inhibitors selective for an alternative form of the core 20S proteosome—the immunoproteasome, which processes peptides for presentation to CD8+ T cells in the MHC-I complex and is constitutively expressed only in hematopoietic cells, induced in immune cells stimulated in the presence of IFN-γ, and upregulated in certain cancers like MM.

Currently, Kezar Life Sciences’ is furthest along in development; in April, it completed a phase 2a trial in autoimmune hepatitis of zetomipzomib (KZ-616), a small-molecule that inhibits both the immunoproteasome core particle component beta subunit 8 (PSMB8; LMP7/β5i) and PSMB9 (LMP2/β1i). Merck kGaA (Darmstadt, Germany) is also pushing forward with a phase 1 clinical program of M3258, a small-molecule inhibitor specific for PSMB8 and intended for use in MM (Principia Biopharma’s selective PSMB8 inhibitor was swallowed up by Sanofi in 2020 when the pharma acquired the San Francisco-based biotech’s Bruton’s tyrosine kinase inhibitor program). Elsewhere, Leiden University startup iProtics recently received a €200K grant from the Dutch Biotech Booster to develop selective immunoproteosome inhibitors, while Auburn University spinout Inhiprot (West Lebanon, NH) received SBIR funding to develop a dual PSMB6/PSMB9 inhibitor for MM. Now, a new study reveals immunoproteosome targeting may also have benefits in neuroinflammatory diseases like multiple sclerosis.

The work, published in Cell and led by Catherine Meyer-Schwesinger and Manuel Friese, from University Medical Center Hamburg-Eppendorf, identifies a neuron-intrinsic mechanism of neurodegeneration in multiple sclerosis (MS) driven by the immunoproteasome.

Under healthy conditions, neurons utilize the constitutive proteasome subunit PSMB5 to regulate proteostasis and degrade 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3), a potent stimulator of glycolysis. This degradation is key because neurons rely more on the pentose phosphate pathway than on glycolysis to produce antioxidants like NADPH and glutathione for protection against oxidative stress.

However, Meyer-Schwesinger, Friese and their colleagues show that, during neuroinflammation, chronic exposure to interferon-γ leads to the induction of the immunoproteasome in neurons, triggering the replacement of constitutive proteosome PSMB5 (β5c) with PSMB8 (β5i). This subunit swap in neurons reduces proteasomal activity, resulting in accumulation of PFKFB3, which in turn enhances glycolysis, diminishes the activity of the pentose phosphate pathway, and impairs redox homeostasis — conditions that sensitize neurons to oxidative injury and ferroptosis.

The team showed that this mechanism was operational in both experimental autoimmune encephalomyelitis (EAE; a mouse model of MS) and brain tissue from MS patients. Moreover, neuron-specific knock-out of Psmb8 or pharmacological inhibition using the small-molecule PSMB8 inhibitor ONX-0914 (originally developed at Onyx Pharmaceuticals/Proteolix) protected neurons in vivo from inflammation-induced damage. Similarly, blocking PFKFB3 with the small-molecule inhibitor PFK-158 or through conditional knockout in neurons reduced disease severity in EAE, prevented neuronal and synaptic loss, and reduced markers of oxidative stress and lipid peroxidation.

It is important to highlight that, unlike cancer or immune cells, neurons do not upregulate PSMB8 in response to a series of MS-related cytokines. So, the neuron-specific effect reported in this study might only become active upon chronic neuroinflammation (i.e. chronic exposure to interferon-γ). Understanding this mechanism might reveal new targets related to the immunoproteosome in the treatment of MS.

This brings us to challenges for translational efforts seeking to develop immunoproteosome inhibitors against MS. Several important neuronal processes, such as synaptic transmission and calcium signaling, are tightly linked to proteasome function; thus, pan-proteosome inhibitors like Velcade could be detrimental to the CNS. The saving grace of approved proteosome inhibitors is that current chemotypes (boronate-based peptides or epoxyketone-based binders) do not cross the blood brain barrier, at least in healthy individuals. Thus, any MS program might need to use intrathecal injection for compounds derived from existing chemical series or engage a medicinal-chemistry effort to design molecules that can breach the BBB and retain potency.

The gambit for immunoproteosome-selective drugs is that they avoid inhibiting constitutive 26S proteosome activity in most tissues (and non-inflammed CNS), which is what makes Velcade and its derivatives so difficult for patients to tolerate; an immunoproteosome inhibitor should therefore have a more favorable safety profile. But so far, immunoproteosome-targeting drugs have had their own share of toxicity problems in the clinic.

Last October, Kezar abandoned its program for zetomipzomib in lupus nephritis after the FDA placed a clinical hold on the trial after 4 patient deaths. The agency placed a second partial hold on the company’s autoimmune hepatitis trial in 24 patients last November due to concerns about steroid control and injection site reactions in 4 patients who were waiting to roll over into the open-label extension arm. Concerns about compromised immune surveillance of acute or latent viral infections due to hobbled antigen processing and presentation would also need to be explored.

In sum, the new work provides strong evidence that the immunoproteosome plays a key role not only in inflammation or infiltration of immune cells, but also in a metabolic switch in neurons which is a key driver of vulnerability in MS. It will be interesting to see whether either targeting immunoproteosome component PSMB8 or taking a completely different tack, blocking PFKFB3, will prove more practical as a neuroprotective strategy in MS.

RESI Boston IPC Winners Announced – Now Accepting Applications for September 

24 Jun

By Max Braht, Director of Business Development, LSN

Max-Braht-HeadshotAt RESI Boston June 2025, 53 companies participated in the Innovator’s Pitch Challenge (IPC), presenting their technologies to panels of active investors and showcasing their work in the Exhibition Hall. RESI attendees — including early-stage investors, startup executives, and industry leaders — voted for the most impressive companies based on pitch presentations, Q&A performance, and networking in the exhibit hall.

Congratulations to the Top-Ranked IPC Companies from RESI Boston June

First Place: MindLab

Our lead product, MLB-001, is a first-in-class, combination drug designed to deliver powerful, life-changing pain relief using significantly lower morphine doses. MLB-001 achieves equal or superior analgesic effects compared to conventional morphine, while minimizing common side effects and reducing the development of drug tolerance. Importantly, MLB-001 is specifically engineered to lower the risk of addiction, a critical advancement given the ongoing opioid crisis. MindLab is committed to redefining pain care by introducing safer, more effective treatments that address unmet medical needs and promote better quality of life for patients worldwide.

Sougato Das, President and COO, Life Science Nation | Larry Raoul James JD, MBA, Founder, President, and CEO of MindLab | Dennis Ford, Founder & CEO, Life Science Nation


Second Place: Inomagen Therapeutics

Inomagen Therapeutics, Inc. is a private, preclinical stage biotechnology company pioneering a gene therapy to improve the treatment of atrial fibrillation. Inomagen has intellectual property and proof of concept data for both the gene and the gene delivery system. Inomagen has a strong and experienced team of industry veterans and key opinion leading cardiovascular physicians to engage in management and advisory roles, including those with extensive domain experience in gene therapy, cardiology, AF therapeutics, medical device, clinical studies, and venture capital. The market size for Inomagen’s gene therapy products is $10.2B.

Sougato Das, President and COO, Life Science Nation | Eric Sandberg, CBO, Inomagen Therapeutics | Robin Drassler, VP of Business Development, Inomagen Therapeutics | Dennis Ford, Founder & CEO, Life Science Nation


Third Place: MantaBio

MantaBio is a life sciences tools company building automated microbial detection systems specifically designed for biopharma manufacturing environments. Our platform offers rapid, high-sensitivity detection across all microbe categories: bacteria, fungi, viruses, and mycoplasma — delivering results in just 2 hours compared to the industry standard of 5 to 28 days. Built with the needs of biologics producers in mind, MantaBio’s technology is optimized for speed, accuracy, and ease of use. It requires less than one minute of hands-on time, no specialized training, and integrates seamlessly into GMP workflows. By enabling at-line or near-line testing with gold-standard sensitivity, MantaBio helps manufacturers reduce contamination risk, minimize batch loss, and accelerate product release timelines. Our system is the next generation in microbial quality control — replacing outdated manual culture methods and reliance on off-site reference labs with a modern, automated approach built to match the pace and complexity of today’s biopharma processes.
Sougato Das, President and COO, Life Science Nation | Max Braht, Director of Business Development, Life Science Nation | Carter Boisfontaine, Co-Founder and President, MantaBio | Dr. Jack Regan, Co-Founder, CEO, and CTO, MantaBio | Claire Jeong, CCO, Vice President of Investor Research, Asia BD, Life Science Nation | Dennis Ford, Founder & CEO, Life Science Nation


We applaud all IPC participants for sharing their innovative work and contributing to a dynamic, high-impact event.

Applications Now Open for the September IPC at RESI Boston

The Innovator’s Pitch Challenge at RESI Boston September offers life science startups the opportunity to present directly to a curated panel of active investors and receive real-time, constructive feedback. Each pitch includes a live Q&A with investor judges and extended exposure through participation in the IPC exhibition hall.

New for September, the IPC package now includes the option to add a second three-day standard ticket at no additional cost. This provides a teammate with full access to partnering and conference sessions.

Benefits include:

  • Investor feedback during live pitch sessions
  • Full access to one-on-one partnering
  • Exhibit table in the IPC Hall
  • Optional additional RESI pass included

Apply now to secure your spot and connect with global early-stage investors this September.

Apply to Pitch at RESI Boston Sept. 2025

The Needle Issue #8

24 Jun
Juan-Carlos-Lopez
Juan Carlos Lopez
Andy-Marshall
Andy Marshall

Around 1 in 5000 people live with a maternally inherited mitochondrial disease like MELASLeber’s Hereditary Optical Neuropathy (LHON) or MIDD, for which there are limited or no treatment options. Gene- and base-editing therapies for mitochondrial DNA (mtDNA) have lagged behind CRISPR–Cas9-based approaches targeting nuclear genes. Whereas there is already a CRISPR–Cas9-based product on the market and >150 different active trials of investigational therapies, the company closest to the clinic with an I-CreI (mitoARCUS) meganuclease targeting a mtDNA point mutation in MELAS/mitochondrial myopathy (Precision Biosciences) announced last month that it was pausing development for commercial reasons.

Despite this disparity, there is reason for optimism as a flurry of different types of optimized cytidine and adenine base editors for mtDNA are now available, with base conversion efficiencies of 50% now achievable, and some newer formats reaching efficiencies as high as 82%.

The development of mtDNA editors is not without challenges. First, editors must dispense with the targeting guide RNA, as mitochondria possess a double membrane that lacks any RNA transport system, effectively thwarting CRISPR-based gene or base editors (instead, a mitochondrial targeting sequence is used to ferry-in editor proteins). Second, unlike nuclear DNA with two copies of a gene, every human cell contains thousands of mitochondria — oocytes contain a whopping 193,000 mitochondria on average — and each organelle contains an average 10 mitochondrial genomes. Those ~10,000 genomes per cell may not all have the same sequence, with mutations existing in a state known as heteroplasmy, in which both mutant and wild-type genomes co-exist in the same organelle. Disease only occurs when the percentage of mutant mtDNA exceeds a particular threshold, typically between 70% and 95%.

Heteroplasmic mitochondrial diseases, like MELAS and MIDD, could be treated using I-Crel/FokI meganucleases or restriction enzymes linked to either transcription activator-like effector (TALE) domains or zinc fingers (which introduce double-strand DNA breaks into target sequences, leading to elimination of mutant mtDNA and repopulation of wild-type mtDNA); other conditions like LHON are predominantly mutant homoplasmic, which means they can only be treated using base editors or supplemental gene therapy.

One key concern with base-editing technology has been its propensity for off-target and bystander changes. This has led to various strategies to increase specificity, such as engineering the deaminases to narrow the editing window or use of nuclear exclusion sequences to stop nuclear sequence editing. Now, two papers in Nature Biotechnology represent important advances that could speed up translational studies of mitochondrial diseases.

Liang Chen, Dali Li and their colleagues of ShanghaiTech University, China report the engineering of highly efficient mitochondrial adenine base editors (eTd-mtABEs) by introducing mutations into the TALE TadA-8e deaminase for greater activity and specificity. These editors achieved up to 87% editing efficiency in human cells and over 50% in vivo, with reduced off-target effects compared to earlier tools.

In the first study, the researchers used eTd-mtABEs to introduce mutations in the human ND6 gene, encoding a subunit of the oxidative phosphorylation (OXPHOS) system linked to LHON and Leigh syndrome. They found reduced levels of ATP and more reactive oxygen species in the edited cells compared with controls, consistent with disease phenotypes. Next, the team used this adenine TALE base editor to introduce two pathogenic T-to-C mutations in the mitochondrial TRNS1 gene of rat zygotes, a gene linked to childhood-onset sensorineural hearing loss. The resulting offspring showed sensorineural hearing loss, which was transmitted to the F1 generation, providing proof of concept that eTd-mtABEs can be used to create animal models of disease.

In the companion paper, Chen, Li and their colleagues used the adenine TALE base editor to model Leigh disease in rats using a similar strategy. The resulting rats showed reduced motor coordination and muscle strength, defects that were obtained with editing efficiencies of only 54% on average. To test if the abnormalities could be reversed, the authors then used a cytosine TALE base editor in zygotes from the mutant rats. On average, the editing efficiency was only 53%, but this was enough to rescue the disease phenotypes.

This is the first report of direct correction of mtDNA mutations via a TALE base editor in an animal model. The next step will be to show feasibility in a model after disease onset (only the UK and Australia allow maternal spindle transfer therapy for mitochondrial diseases; no country has permitted mitochondrial base editing in human zygotes).

Achieving effective therapeutic mitochondrial base editing in affected target tissues will thus require efficient AAV delivery. For LHON, an already approved FDA AAV-2 product transduces the optic nerve and retinal ganglion cells, providing a translational path; GenSight Biologics also recently published 5-year outcome data for its AAV-2 therapy Lumevoq (lenadogene nolparvec) in LHON. But AAV delivery in other mitochondrial conditions will not be as simple: MELAS patients, for example, require efficient transduction of the CNS, kidney, skeletal muscle and cardiac muscle; MIDD patients need AAV delivery to the pancreas, inner ear, retina and kidney. Although a commercial AAV vector (AAVrh74) is available for muscle (Sarepta’s Elvidys), vectors that reach many of these other tissues have yet to be commercialized and may require next-generation AAV capsids and/or refinement of machine-guided design of cell type-specific synthetic promoters to reach target organs.

It is encouraging that the roughly 50% base conversion rate achieved in these new studies exceeded the heteroplasmy threshold required for disease manifestation and therapeutic rescue. At the same time, despite this remarkable success, concerns remain about off-target effects — both in mitochondrial and nuclear genomes — and narrow therapeutic windows. And with base editing approaches so far behind conventional gene therapies like Lumevoq in development, compelling commercial and clinical advantages benchmarked against best-in-class gene therapy will be needed to convince investors to back these approaches.

One parting thought: the past year has seen a noticeable uptick in publications on mitochondrial base editing technology from labs outside of the US. TALEN specialist Cellectis, headquartered in Paris, France, acquired 19% of equity in the mitochondrial base editing company Primera Therapeutics in 2022, ostensibly for its rapid TALE assembly platform (FusX System), which streamlines TALE repeat construction. In South Korea, Jin-Soo Kim at the Korea Advanced Institute of Science and Technology (KAIST) recently co-founded startup Edgene with Myriad Partners to develop mitochondrial base editors based on his seminal work on TALE-linked deaminases (TALEDs) enabling A to G conversion, which he has continued to optimize. According to Biocentury8 out of 13 base editing studies published in 27 translational journals over the past year came from labs in China. Wensheng Wei’s group at Peking University, a founder of Edigene in Beijing, continues to work on mitobase editors, with two recent patents on strand-selective mitochondrial editing. And Jia Chen of ShanghaiTech University, China, and his collaborators Li Yang and Bei Yang, are scientific advisors to Correctseq in Shanghai, which is developing transformer base editors for ex vivo and in vivo applications. It seems that mitochondrial base editing may be another area where US biotech may soon be finding itself chasing the dragon. David Liu and Beam Therapeutics may have something to say about that.

The Needle Issue #7

10 Jun
Juan-Carlos-Lopez
Juan Carlos Lopez
Andy-Marshall
Andy Marshall

Ex vivo HSC lentiviral gene therapies have been on the market for nearly a decade, with six products approved and at least 55 now in clinical testing for rare inherited diseases, HIV infection or cancer. And yet, their commercial success remains in question. Bluebird Bio—which was valued at $10 billion only a few years ago and successfully shepherded to market Zynteglo against transfusion-dependent β-thalassemia, Skysona for early cerebral adrenoleukodystrophy, and Lyfgenia for sickle-cell disease (SCD)—was sold earlier this year to private-equity firms Carlyle and SK Capital for a measly $29 million. Last November, the company had treated only 57 patients (35 for Zynteglo; 17 for Lyfgenia and 5 for Skysona), with just 28 of 70 medical centers across the US ready to treat patients due to delays in accreditation and training of personnel. In Europe, Orchard Therapeutics halted marketing and production of a treatment for severe combined immunodeficiency caused by adenosine deaminase mutations (Strimvelis) after six years, forcing Fondazione Telethon to take over production. Even market uptake of Vertex’s much-heralded CRISPR/Cas9 BCL11a SCD therapy Casgevy has been sluggish.

These subpar commercial launches relate to the complexity of ex vivo lentiviral gene therapy: patient identification and qualification is lengthy; HSC mobilization and sourcing efficiencies vary due to patient heterogeneity; and manufacture and distribution processes remain lengthy and convoluted (sometimes requiring repetition if a poor quality product batch is generated). From first evaluation, patients are required to make several hospital visits over a period (of up to a year) and must undergo punishing conditioning regimes with lymphodepletive bisulfan before infusion, which itself carries infertility and cancer risks. All of these challenges have added impetus to the search for alternative and more efficient approaches for carrying out HSC gene therapy.

A group led by Alessio Cantore and Luigi Naldini, from the San Raffaele Telethon Institute for Gene Therapy in Milan, Italy, report in Nature that it may be possible to obviate these challenges by delivering recombinant lentiviral vectors in vivo soon after birth, when HSCs continue to circulate in the bloodstream in large numbers and are beginning their transition from the liver (where they are located in the fetus) to bone marrow (where they remain through adulthood).

Cantore, Naldini and their colleagues started by measuring the number of circulating HSCs in neonatal, 1-, 2- and 8-week-old mice, looking at the peripheral blood, spleen, liver and bone marrow. They found that HSCs were present in the circulation right after birth and that their number immediately declined. These cells could be transduced with lentiviruses, successfully engrafted, and persisted in the mice for several months.

To show that these HSCs could be harnessed to treat genetic disorders, the team tried to correct three mouse models of disease — adenosine deaminase deficiency, autosomal recessive osteopetrosis and Fanconi anemia. Although the therapeutic effect of the cells varied depending on the disease, the results provided compelling evidence for the potential for in vivo gene transfer to HSCs.

The authors reported that human neonates also have circulating HSCs in high numbers. And although the therapeutic window in the mouse only existed during the neonatal period, it was possible to lengthen it by mobilizing the HSCs from their niche in two-week-old animals using protocols in clinical use (granulocyte-colony stimulating factor/CXCR4 antagonist Plerixafor) These observations raise the possibility of therapeutically targeting HSCs in newborns, potentially opening the gates to treatment of a variety of inherited conditions.

Compared with the headaches of ex vivo manipulation, the authors’ concept of simply injecting a lentiviral gene therapy into a newborn to bring about a genetic cure is certainly alluring. But reducing this to clinical practice will require optimization of many different factors. How to account for the heterogeneity and fragility of patient HSCs in a particular disease? How to measure the cellular activation/metabolic state of HSCs in newborns and assess the affect on amenability to lentiviral transduction in the hostile milieu of blood? What effect would shear stress in circulation have on lentiviral transduction efficiencies in situ? What would be the selective engraftment advantage provided to HSCs after engraftment of a particular gene? And what would be the potential safety implications of off-target transduction events in cells other than HSCs, given instances of dysplastic syndromes have been reported with ex vivo lentivectors?

Current ex vivo lentiviral gene therapy like Lyfgenia and Zynteglo infuse between 3–5×106 gene-modified CD34+ HSCs/kg in a patient. The challenge for in vivo lentiviral gene therapy will be to achieve transduction efficiencies that transduce as many cells and obtain similar engraftment rates in the rapidly turning over HSC population. Beyond these issues, there are additional practical challenges: can genetic testing of an infant happen fast enough to take advantage of the short therapeutic window for which an in vivo lentiviral HSC therapy could work?

Clearly, the new work raises many intriguing questions for the lentiviral gene therapy space. And for newborns with genetic diseases, such as severe immunodeficiencies or Fanconi anemia, in vivo HSC gene therapy may open up new treatment options.