Development At The Speed Of Light: Pharma's Search For A COVID-19 Vaccine

 
CONTINUE READING
Development At The Speed Of Light: Pharma's Search For A COVID-19 Vaccine
Development At
The Speed Of Light:
Pharma’s Search For A COVID-19 Vaccine
Development At The Speed Of Light: Pharma's Search For A COVID-19 Vaccine
Agenda
  Vaccine landscape and frontrunners
  Pipeline snapshot
  Who are the frontrunners?
  Potential launch timelines and upcoming catalysts
  Clinical data – what do we know and what questions remain?

  Regulatory hurdles
  Compressed clinical development timelines
  FDA guidance maintains high standards for pivotal efficacy trials
  Enrolling clinically-relevant populations
  Optimal clinical trial site selection

 Commercialization challenges
 How large will vaccine demand be?
 Can manufacturing capacity meet demand? (active ingredient + fill-finish)
 How can equitable access to a limited supply of doses be achieved?
 Pricing dilemma – how to profit without impairing market access?

                                                                             2
Development At The Speed Of Light: Pharma's Search For A COVID-19 Vaccine
Vaccine landscape
Development At The Speed Of Light: Pharma's Search For A COVID-19 Vaccine
Explosion in industry-sponsored trials as developers race for
approval
121 planned or ongoing trials for preventative and therapeutic vaccines

 Source: Trialtrove, 17 July 2020                                         informa | Pharma Intelligence   4
Development At The Speed Of Light: Pharma's Search For A COVID-19 Vaccine
Unprecedented interest in vaccine development from
both established and newcomer companies
 168 pipeline vaccines
 22 vaccines in clinical development                     *Showing top 25 companies
 6 technologies - inactivated, viral vector, DNA, RNA,
  recombinant protein, virus-like particle (VLP)

 Source: Pharmaprojects, 17 July 2020                                       informa | Pharma Intelligence   5
Development At The Speed Of Light: Pharma's Search For A COVID-19 Vaccine
Public funding and partnerships have provided an opportunity
for smaller players to evaluate new technologies
Vaccine                              Company                               Phase   Technology               Public funding
AZD1222                              University of                         III     ChAdOx1 vector vaccine   £65.5m from UK government and up to $1.2bn
                                     Oxford/Vaccitech/AstraZeneca                                           from BARDA

Sinopharm inactivated vaccine        Sinopharm (China National             III     Inactivated vaccine      China state-owned
                                     Pharmaceutical Group Corp)
PiCoVacc                             Sinovac                               II      Inactivated vaccine      $8.5m from Chinese government

mRNA-1273                            Moderna                               II      mRNA vaccine             $483m from BARDA

BNT162                               BioNTech/Pfizer/Fosun Pharma          II      mRNA vaccine             €100m from European Investment Bank (Pfizer
                                                                                                            has declined US funding)
Ad5-nCoV                             CanSino Biologics/Beijing Institute   II      Ad5 vector vaccine       R&D support from Chinese Academy of Military
                                     of Biotechnology                                                       Medical Sciences

NVX-CoV2373                          Novavax                               I/II    Recombinant protein      $1.6bn from US government and $384m from
                                                                                   nanoparticle vaccine     CEPI
INO-4800                             Inovio                                I/II    DNA vaccine              $71m from US Department of Defense and
                                                                                                            $17.2m from CEPI
Trimer-Tag vaccine                   Clover/GSK/Dynavax                    I       Spike trimer subunit     $69.5m from CEPI
                                                                                   vaccine + adjuvant
CureVac mRNA vaccine                 CureVac                               I       mRNA vaccine             €100m from European Investment Bank and
                                                                                                            €300m from German government
Medicago VLP vaccine                 Medicago/GSK/Dynavax                  I       VLP                      Undisclosed funding from Canadian government
Imperial College London vaccine      Imperial College London               I       Self-amplifying mRNA     £41m from UK government
                                                                                   vaccine
     Source: Datamonitor Healthcare, July 2020
                                                                                                                                             informa | Pharma Intelligence   6
Development At The Speed Of Light: Pharma's Search For A COVID-19 Vaccine
Who are the
frontrunners?
Development At The Speed Of Light: Pharma's Search For A COVID-19 Vaccine
Front runners with potential 2020 Emergency Use Authorization
                                                              Phase I/II

                                                              Phase II/III

                                                     Possible EUA in October 2020
                                                     (September in UK)

                                                     Possible EUA in October 2020

                                                     Possible EUA in November 2020

                                                     Possible EUA by year end

                                                     Possible EUA by year end

                                                     Possible EUA by year end

                                                     Possible EUA by year end

   Source: Datamonitor Healthcare, July 2020
                                                                    informa | Pharma Intelligence   8
Development At The Speed Of Light: Pharma's Search For A COVID-19 Vaccine
Later market entrants will play crucial role in boosting supply

Vaccine                                      Phase         Catalyst
NVX-CoV2373                                  I/II          Phase I immunogenicity data expected in July. Phase III to
(Novavax)                                                  initiate in Autumn, utilizing Warp Speed funds
Ad26.COV2-S                                  Preclinical   Phase I/II trial initiating in July with Phase III targeted for
(Johnson & Johnson)                                        September 2020 and EUA in Q1 2021
Adjuvanted subunit vaccine (Sanofi/GSK)      Preclinical   Phase I expected in H2 2020, anticipated approval in H2
                                                           2021
Measles vector-based vaccine                 Preclinical   Phase I trials in H2 2020
(Merck/Themis)
VSV vector-based vaccine
(Merck/IAVI)

 Novavax, J&J, and Merck & Co are part of the “Operation Warp Speed” program. All three have
  received US-government funding to fund clinical development and scale up manufacturing capacity
 Novavax is targeting 100m doses by the end of 2020, and up to 1bn dose by the end of 2021
 J&J is also targeting 1bn doses by the end of 2021 with a single-dose schedule
 Merck & Co is ultimately targeting 1bn doses, and replicative nature of viral vectors may facilitate
  single-dose schedules

 Source: Datamonitor Healthcare, July 2020
                                                                                                                     informa | Pharma Intelligence   9
Development At The Speed Of Light: Pharma's Search For A COVID-19 Vaccine
Phase I insights and
remaining questions
mRNA vaccines have shown great promise in Phase I
Compelling immunogenicity as measured by neutralizing antibody titers

 BNT162b1 (Pfizer/BioNTech) which encodes S protein receptor-binding domain, and mRNA-1273 (Moderna) which encodes
  full S protein, have both demonstrated ability to induce neutralizing antibody titers that were comparable to or exceeded
  those observed in sera of convalescent patients
 Caveat is that serum panels in both studies were primarily from patients with mild-moderate disease, who have lower
  antibody titers. Pfizer/BioNTech panel may also not have been at peak of immune response (“at least 14 days after
  confirmed diagnosis”) and included individuals older than those who were vaccinated (18-83 years vs. 19-54 years,
  respectively). Moderna panel were 23-60 days post-symptom onset, age not specified.
 Moderna also released data on cellular immunogenicity showing Th1-biased CD4 T-cell responses without significant
  elevation of Th2-biased CD4 T-cell responses
 Cellular immunogenicity data are expected from ongoing BioNTech European study, as well as additional data on the three
  other constructs being evaluated.
                                                                                         BNT162 constructs under evaluation

 Vaccine (Phase III               Vaccine GMTs               Convalescent
 dose)                                                       sera Nab GMTs
 BNT162b1 (30ug)                  267                        94
                                  (Day 28; PRNT50)
 mRNA-1273 (100ug)                654 (Day 43, PRNT80)       158 (n=3)
                                  344 (Day 57, PsVNA ID50)   109 (n=38)

  Source: BioNTech, July 2020; Moderna, July 2020
                                                                                                                  informa | Pharma Intelligence   11
Key remaining questions:
Will antibodies/T-cell responses confer protection?
 Correlates of protection are still unknown meaning there is no guarantee that immunogenicity will translate into protective
  efficacy in pivotal studies

How durable are neutralizing antibody responses?
 Follow-up has been limited to one month post-vaccination thus far. If responses are not durable, intra-season protection
  could be compromised and seasonal vaccination may be required

Will similar immunogenicity be observed in elderly, ethnic minorities, and patients with co-morbidities?

 Both studies were limited to healthy adults aged 18-55 years, but the elderly is a key risk group and immune responses tend
  to be lower due to immunosenescence. Future pivotal studies aim to enrol diverse patient groups considered at high-risk

Reactogenicity a potential weakness?
 Severe fatigue and severe chills occurred in one patient each in BNT162 30ug arm (post-2nd dose), and severe erythema was observed
  in 1 patient in mRNA-1273 100ug arm (post-2nd dose). Highest doses discontinued due to high frequency of severe adverse events.
 Not a barrier to approval, but could emerge as a weakness if other approaches are less reactogenic. We note that all adverse events in
  Inovio’s Phase I trial (INO-4800, n=40) were mild.

  Source: Datamonitor Healthcare
                                                                                                                informa | Pharma Intelligence   12
Mixed data for adenoviral vector vaccines thus far
CanSino Ad5 vaccine immunogenicity impaired by pre-existing anti-vector antibodies
 Disappointingly low rates of seroconversion for neutralizing antibodies to live SARS-CoV-2 observed in 108 healthy adults
  aged 18-60 years, though T-cell responses were observed
 Single dose administered at three dosing levels (5x1010 vp/ml, 1x1011 vp/ml, 1.5x1011 vp/ml)
 Pre-existing anti-vector antibodies compromised immunogenicity of the vaccine. In addition, recipients aged 45-60 years had
  lower neutralizing antibody titers than younger participants.

    Neutralizing antibodies (live                    Low dose (n=36)   Middle dose (n=36)   High dose (n=36)
    SARS-CoV-2 assay)
    GMTs (day 28)                                    14.5              16.2                 34.0

    ≥4-fold increase (day 28)                        18 (50%)          18 (50%)             27 (75%)

 Highest dose (1.5x1011 vp/ml) discontinued due to higher frequency of severe adverse events (17%)

Positive Phase I data for AZD1222, but preclinical model suggests it may only provide partial protection
 Phase I data from 500 subjects showed stimulation of both humoral and cellular immunity after a single dose
 However, preclinical challenge study in six rhesus macaques showed that the vaccine did not prevent infection, but did provide
  protection from pneumonia compared to unvaccinated controls. No difference in viral loads in nose swabs also suggests vaccine
  will not prevent individuals transmitting the virus, which has implications for efforts to achieve herd immunity
  Source: Doremalen et al., 2020; Zhu et al., 2020
                                                                                                               informa | Pharma Intelligence   13
Regulatory
requirements
Some helpful definitions for the next slide…
Probability of Success (PoS): The probability of successfully advancing from one clinical stage to another, or
from NDA/BLA to approval.

PoS is expressed as a percentage. It represents the percentage of products that successfully transitioned
through a phase, divided by the number of products in that Phase that have undergone a Phase transition
(either successfully or unsuccessfully). For example, if there were 100 drugs in Phase II development and 50
transitioned to Phase III, 20 were suspended and 30 remained in Phase II development, the probability of
success would be:
50/70 = 71.4%.

Likelihood of Approval (LOA): The probability of a drug gaining FDA regulatory approval from its current phase
of development.

For example, if a drug is currently in Phase II, and the probability of success for Phase II is 30%, Phase
III is 50%, and NDA/BLA is 80%, then the likelihood of approval for the Phase II drug would be:
(30%*50%*80%) = 12%

Source: Pharmapremia, July 2020                                                                         informa | Pharma Intelligence   15
Urgency has led to unprecedented compression of clinical
development timelines

 Average clinical development duration for vaccines is 11 years
 Average likelihood of approval for a a Phase I vaccine is 16.5%
 Pfizer/BioNTech, Moderna, CanSino, and AstraZeneca/University of Oxford have all progressed from Phase I to Phase III
  within 2–4 months, with fastest possible approval within a 5-month clinical development timeframe (Pfizer/BioNTech)

 BUT… regulators are rightly insisting on large-scale trials, in at-risk patients, with robust efficacy and safety endpoints

  Source: Pharmapremia, July 2020                                                                        informa | Pharma Intelligence   16
FDA guidance sets a rigorous standard for clinical trial design

    “There are currently no accepted surrogate endpoints that are reasonably likely to predict clinical benefit
    of a COVID-19 vaccine. Thus, at this time, the goal of development programs should be to pursue
    traditional approval via direct evidence of vaccine safety and efficacy in protecting humans from SARS-
    CoV-2 infection and/or clinical disease.”

 Key requirements/recommendations:
 Trial size recommendations: Several thousand participants required to have reasonable probability of demonstrating protective
  efficacy. Moderna/AstraZeneca/Pfizer pivotal trials are targeting 30,000 patients each
 Must enroll ‘at-risk’ patients: This includes the elderly, ethnic minorities, and patients with relevant co-morbidities. This is
  essential for both clinical relevance and to maximize the probability of demonstrating efficacy
 Acceptable efficacy endpoints: Either laboratory-confirmed COVID-19 or laboratory-confirmed SARS-CoV-2 infection is an
  acceptable primary endpoint. Reduction in severe COVID-19 disease should be evaluated as a secondary endpoint (if not a
  primary endpoint), and incidence of both symptomatic and asymptomatic infections should be evaluated.
 Required safety endpoints: Solicited local and systemic AEs for at least 7 days after vaccination, unsolicited AEs for at least 21–
  28 days after vaccination, and serious and other medically attended AEs for at least 6 months after final dose
 Follow-up: At least 1–2 years to gauge longevity and potential for vaccine-associated enhanced respiratory disease
 Comparator: Placebo control arm is necessary, but active comparators may be possible in future

 Source: FDA, June 2020                                                                                     informa | Pharma Intelligence   17
Choice of clinical site locations must reflect changing
epicenter of the pandemic
14-day cumulative reported COVID-19 cases per 100,000

                                                         Trial site selection is crucial to
                                                          ensure a high enough COVID-19
                                                          attack rate in the placebo arm to
                                                          be able to demonstrate vaccines
                                                          have statistically significant
                                                          protective efficacy

                                                         Brazil, Chile, Russia, South Africa,
                                                          Saudi Arabia, UAE, and US are sites
                                                          in announced pivotal trials

Source: ECDC, July 2020                                                  informa | Pharma Intelligence   18
Key Commercialization
Challenges
Key challenges:
How large will vaccine demand be?
 Which risk groups are likely to be prioritized? WHO suggests 24% of global population (~1.9bn) fall into highest priority groups
  (healthcare workers, elderly, and individuals with certain co-morbidities such as diabetes and hypertension)
 What coverage rates are required for herd immunity? What coverage can be realistically achieved? Will public perception be
  affected by rushed development process and declining case numbers in some markets?
Can manufacturing capacity meet demand (both active ingredient and finish-fill capacity)?
 Significant CEPI and BARDA investments have allowed manufacturers to partially de-risk scale up process ahead of demonstrating
  proof-of-concept
 Competition for finish-fill capacity could be a major hurdle. Manufacturing demand for therapeutics must also be considered
How can equitable access to a limited supply of doses be achieved?
 Nationalistic agreements with manufacturers to secure supplies in advance will inevitably result in lower-income markets losing out
 Supranational national initiatives such as COVAX (ACT-Accelerator vaccines pillar) which aims to secure 2bn doses by 2021, and the EU
  Emergency Support Instrument will play crucial roles in ensuring equitable access, though the former is still far off fundraising target
  ($18.1bn)
What is an appropriate price to balance need for profitability with ensuring market access?
   Traditional HTA processes are too slow and required data may not be available, thus short-term price agreements will be necessary
   Prices must reflect substantial public investment in the R&D process and lesser ability to pay of lower-income markets
   Initial altruistic approaches of certain companies could act to set short-term price cap for rivals with greater focus on profit
   Possible seasonal recurrence/annual vaccination has implications for longer-term profitability

    Source: UN Population Prospects, 2019; WHO, 2020
                                                                                                                    informa | Pharma Intelligence   20
Vaccine supply estimates by end of 2021 = 8.3bn doses*

                                                           *Figure only includes
                                                           estimates from
                                                           companies which have
                                                           explicitly stated a
                                                           targeted dose capacity
                                                           in 2021

Source: Company statements
                                                         informa | Pharma Intelligence   21
Thank you
Questions?
Email: pharma@informa.com
You can also read