Skip to Main Content

CII - David Martinez, PhD

August 28, 2023
  • 00:00So first, David Martinez.
  • 00:02He joined the immunobiology
  • 00:05department just three months ago,
  • 00:07so welcome to Yale.
  • 00:10So David grew up in El Salvador
  • 00:12and he came to the United States,
  • 00:14where he had the opportunity to work
  • 00:17with very prominent scientists.
  • 00:18So Sally Permars, who is a prominent
  • 00:22immunologist and vaccinologist.
  • 00:24She was his advisor during his doctoral
  • 00:27studies at Duke University and then he
  • 00:30joined Ralph Barrick's lab at UNC Chapel
  • 00:33Hill from his postdoctoral studies.
  • 00:35And David's core expertise
  • 00:38is vaccinology and virology,
  • 00:40focusing focusing on inflated
  • 00:43viruses and coronaviruses.
  • 00:44And I wanted to highlight that
  • 00:46not only has he made very
  • 00:48interesting observations for dengue,
  • 00:50but he also has been very involved
  • 00:54throughout the pandemic and
  • 00:55he helps to understand.
  • 00:57So to inform on the development
  • 00:59of the vaccines,
  • 01:01the Moderna and the J&J,
  • 01:03as well as the antibody,
  • 01:05the antivirals from Eli,
  • 01:06Lilly and AstraZeneca,
  • 01:08which is really impressive.
  • 01:09He's also a Hannah Gray,
  • 01:11faculty fellow of the HTMI.
  • 01:13And so please join me in welcoming
  • 01:15David for his first talk at Yale as
  • 01:17a tenure track assistant professor.
  • 01:37All right, Well, thank you very much,
  • 01:38Nicole, for that very kind introduction.
  • 01:41Thank you all for joining the launch of the
  • 01:43Yale Center for Infection and Immunity.
  • 01:46I'm incredibly excited to be here
  • 01:48enjoying such an outstanding but also
  • 01:51incredibly friendly and supportive group.
  • 01:54Also, thank you to the Dean for
  • 01:56the support of the Center for
  • 01:58Infection and Immunity and as well,
  • 02:03Nicole, for planning this and and
  • 02:05and just getting this set up.
  • 02:07So I'd like to tell you a little bit
  • 02:10about some of the approaches that we've
  • 02:12been thinking about over the last really
  • 02:143 1/2 years in terms of tackling these
  • 02:17preemergent and pandemic coronaviruses.
  • 02:22So this is I think, information
  • 02:24that's hopefully familiar by now.
  • 02:27But you know, we really have around,
  • 02:29depending on who,
  • 02:30how we define viral families, 24 to 26
  • 02:33viral families that can infect humans.
  • 02:35This is one of those viral families that
  • 02:38includes members of the family Corona Verde.
  • 02:41And from these family we really have
  • 02:44a four genera that exists in in here.
  • 02:46And from these four genera,
  • 02:48two of them are more common
  • 02:50commonly infecting humans.
  • 02:51So these include the alpha coronaviruses
  • 02:53that are shown here in in this cluster,
  • 02:57as well as the beta coronaviruses that
  • 02:59are shown here by these pink clusters
  • 03:02from the beta coronavirus genus.
  • 03:05We really have two major groups
  • 03:07that are known to infect humans.
  • 03:09These include the group to be
  • 03:11sarbecoviruses or SARS related viruses,
  • 03:13which of course include members of
  • 03:15that we're familiar with including
  • 03:16SARS COVID One which emerged in
  • 03:182003 and SARS COVID 2A virus
  • 03:21that now needs no introduction.
  • 03:23We also have the group to see Merbeka
  • 03:25viruses or MERS related viruses.
  • 03:27So really it wasn't until the
  • 03:29early 2000s that people began to
  • 03:31pay attention to these groups.
  • 03:33Whenever SARS coronavirus emerged
  • 03:35and infected over 8000 people
  • 03:38and caused around 800 deaths,
  • 03:40so about 10% mortality rate.
  • 03:43Some of these other viruses
  • 03:44are more pathogenic,
  • 03:45including MERS coronavirus.
  • 03:46It's estimated to cause around
  • 03:49a 30% mortality rate.
  • 03:50And the reasons underlying the higher
  • 03:53mortality rate of the virus are unknown,
  • 03:56although it's speculated that
  • 03:57some of the genes and the virus
  • 04:00itself and the accessory orbs
  • 04:02are potentially responsible for
  • 04:03this increased pathogenesis.
  • 04:05So in thinking about how to tackle
  • 04:07these genetically divergent viruses,
  • 04:09we began to look at one of these
  • 04:14subgenera of the Sarbeca viruses.
  • 04:16So these include the zoonotic,
  • 04:17SARS like viruses.
  • 04:19And this similarity plotter,
  • 04:21still called Simplot,
  • 04:22really depicts the genetic
  • 04:24diversity within this subgenus of
  • 04:27viruses throughout its genome.
  • 04:29So these are fairly large RNA viruses,
  • 04:31around 30 KB depending on
  • 04:33which virus we're looking at.
  • 04:35And these viruses on the right here
  • 04:38are zoonotic viruses of bad origin,
  • 04:40as well as human epidemic viruses
  • 04:43or other zoonotic viruses.
  • 04:44And what I want to draw your attention
  • 04:46to in this particular similarity
  • 04:48plot is that these viruses have a
  • 04:50great degree of genetic diversity in
  • 04:52various portions of their genomes,
  • 04:53in particular the subunit one that is
  • 04:57depicted here by the spike that I've laid,
  • 04:59this ribbon model that have
  • 05:02laid sort of parallel to this.
  • 05:04So when we began to think about how
  • 05:06do we begin to tackle these viruses
  • 05:09that are genetically diverse,
  • 05:10how are we going to deal with the
  • 05:12genetic diversity of these viruses?
  • 05:13So this depicted the bigger the dip,
  • 05:15then the more genetic diversity
  • 05:17we have among these viruses.
  • 05:19So to begin to do this,
  • 05:21we turn to patients that have
  • 05:23been infected with the original
  • 05:24SARS COVID from 2003.
  • 05:26So this was a highly collaborative study
  • 05:29spanning the NIAID Vaccine Research Center,
  • 05:34UNC,
  • 05:35Duke University and and other collaborators.
  • 05:37So what we did is we got a hold of
  • 05:41a convalescent PBMC samples and we
  • 05:43began to study monoclonal antibodies
  • 05:45from these from this particular patient.
  • 05:47So this was a patient that I,
  • 05:49like I said had been infected with
  • 05:50SARS COVID in 2003 and by standard
  • 05:54approaches that have been developed
  • 05:56by others and also utilized by
  • 05:58us and many others.
  • 05:59Then we began to isolate antigen
  • 06:02specific memory B cells by essentially
  • 06:05taking an antigen bait protein,
  • 06:08labeling it and then sorting
  • 06:09those memory B cells and cloning
  • 06:11out monoclonal antibodies.
  • 06:12And we began to triage the antibodies
  • 06:15and study them in a way that we could
  • 06:17begin to map to where they bound on
  • 06:19the spike protein on the SARS Kobe 2 virus,
  • 06:22but also other SARS related viruses.
  • 06:25And I'm going to tell you a little
  • 06:27bit about those monoclonal antibodies.
  • 06:29But first I want to just for the,
  • 06:31so that my talk is clear,
  • 06:32just want to introduce this other concept.
  • 06:34So I think this is something that
  • 06:36we're all familiar with by now just
  • 06:38you know given what we've heard in
  • 06:39the media over the last few years
  • 06:41about antibodies to the coronavirus.
  • 06:42But I think it's an important
  • 06:44concept just for clarity of my talk.
  • 06:47So antibodies have multiple functions.
  • 06:49They can have antiviral functions
  • 06:52like nonneutralizing functions like a
  • 06:55BCC or various phagocytic functions.
  • 06:57But one of the functions that they
  • 06:59do not on the nonneutralizing side,
  • 07:01but the other function is neutralization.
  • 07:04So this is the literal ability of
  • 07:05the Mal antibody molecule to stop
  • 07:07the virus from delivering its its
  • 07:09genetic cargo inside the cell.
  • 07:11So for a lot of the data that I'm going
  • 07:13to show you, when I say neutralization,
  • 07:15I'm literally talking about this.
  • 07:17So antibodies can neutralize by essentially
  • 07:19stopping the conformational change of
  • 07:21the spike protein which is, you know,
  • 07:23essentially a spring loaded molecule.
  • 07:25And in doing so,
  • 07:26it can prevent the delivery of the viral
  • 07:29cargo that the genetic cargo into the cell.
  • 07:31So we screened over 1700 monoclonal
  • 07:33antibodies that were isolated
  • 07:35from this particular patient.
  • 07:37And I don't have time to talk about
  • 07:38all the 1700 monoclonal antibodies,
  • 07:40but I want to draw your attention to
  • 07:42four of these monoclonal antibodies
  • 07:43that really caught our attention.
  • 07:45So these were antibodies that could
  • 07:47neutralize various SARS related viruses,
  • 07:49including viruses of zoonotic origin
  • 07:52from bat species as well as the
  • 07:54SARS Co V2 pandemic virus and the
  • 07:57original SARS coronavirus from 2003.
  • 08:00So from these four monoclonal antibodies,
  • 08:03what this data told us is that
  • 08:05despite the genetic diversity within
  • 08:06the subunit 1 portion of the spike,
  • 08:08then clearly there were sites that were
  • 08:11so-called Achilles heels within these
  • 08:12viruses that could be targeted by the
  • 08:14host immune response and in particular,
  • 08:16the human immune response,
  • 08:18which was not really known at the time.
  • 08:21So we began to study these
  • 08:23monoclonal antibodies,
  • 08:23not just in vitro but also in vivo.
  • 08:25So for for much of the data that I'm going
  • 08:27to show you in this portion of my talk,
  • 08:29then it really involves characterizing
  • 08:31the antiviral functions of these
  • 08:34monoclonal antibodies in mouse models.
  • 08:36And just to give you a brief overview
  • 08:39of how the experiments are being set up
  • 08:41and they're essentially two different
  • 08:42ways that we evaluated these antibodies,
  • 08:44either in a prophylactic or preventative
  • 08:47setting or in a therapeutic setting.
  • 08:49So in these various animal models
  • 08:52of coronavirus pathogenesis,
  • 08:53then peak virus replication is around
  • 08:562 days post infection or 48 hours.
  • 08:58So we want to evaluate the antibodies
  • 09:00in a manner that we can test whether or
  • 09:03not viral replication and the relevant
  • 09:06tissues of these mice like the lung or
  • 09:08the upper Airways can in fact be dampen.
  • 09:10So we either treated the mice at 12
  • 09:13or or 12 hours before infection or 12
  • 09:15hours post infection or even at times
  • 09:18later post infection to determine
  • 09:19whether or not the antibodies had any
  • 09:22efficacy against these various viruses.
  • 09:25So looking at this data,
  • 09:27we first began to evaluate the
  • 09:30antiviral function of four of these,
  • 09:32you know,
  • 09:32broadly neutralizing crossreactive
  • 09:34antibodies against the original SARS.
  • 09:36So this is SARS Co V1 and a highly
  • 09:39pathogenic mouse model.
  • 09:41So from this data,
  • 09:42whenever we compared the ability
  • 09:44of these antibodies to block virus
  • 09:46replication in the lungs of these
  • 09:49animals compared to a negative
  • 09:50controlled flu monoclonal antibody,
  • 09:52then only one of these antibodies was really
  • 09:56able to fully suppress virus replication.
  • 09:58So for the remainder of this
  • 09:59particular part of my talk that I'm
  • 10:01going to focus on telling you about
  • 10:02that particular monoclonal antibody,
  • 10:04which is called DH1047.
  • 10:05So we wanted to know whether
  • 10:07or not this antibody could not
  • 10:09only prevent disease in these mice,
  • 10:11but could it also dampen virus
  • 10:13replication in a therapeutic setting?
  • 10:15So once the mice are infected,
  • 10:16if we treat them with
  • 10:18this particular antibody,
  • 10:19can we mitigate signs of viral pathogenesis?
  • 10:22And that is in fact what we saw
  • 10:25whenever mice were treated with
  • 10:27DH1047 in a therapeutic setting,
  • 10:29then we could reduce lung virus
  • 10:31replication as well as other
  • 10:33measures of viral pathogenesis.
  • 10:36We then wanted to expand our breath
  • 10:39of studying this monoclonal antibody
  • 10:41and also evaluate the ability of this
  • 10:43particular antibody isolated from
  • 10:45a human and blocking these various
  • 10:48coronaviruses and mouse animal models.
  • 10:50So we looked at A2 zoonotic
  • 10:53preemergent viruses that I'll note
  • 10:56that whenever you take these two
  • 10:58coronaviruses that are shown here,
  • 11:00they're isolated from a bat and
  • 11:02you put them in human primary
  • 11:03airway epithelial cells,
  • 11:05they grow efficiently well and they
  • 11:07can evade many of the commonly used
  • 11:09countermeasures at least against SARS
  • 11:11COVID and and also against SARS,
  • 11:13COVID too.
  • 11:14So but this,
  • 11:15this monoclonal antibody that we
  • 11:17isolated from a human then could not
  • 11:19only prevent the virus infection
  • 11:21and pretreated mice,
  • 11:22but could also treat the virus
  • 11:24infection in these mice.
  • 11:25Suggesting that within these
  • 11:27highly genetically diverse subunit
  • 11:291 portion of the spike then this
  • 11:31was again pointing to an Achilles
  • 11:34heel within these various viruses.
  • 11:36And then also in looking at again
  • 11:38looking at more breadth of this
  • 11:40virus as we also evaluated another
  • 11:43SARS related virus called WIV 16.
  • 11:45And then we could similarly treat
  • 11:48these mice in a preventative or or
  • 11:50therapeutic setting and also against
  • 11:52a SARS two related virus that was
  • 11:54isolated from an animal called a pangolin.
  • 11:57So clearly the,
  • 11:59the footprint bound by this
  • 12:01monoclonal antibody then it's highly
  • 12:03must be highly conserved.
  • 12:04So we were interested in also
  • 12:06testing the ability of this antibody
  • 12:08against the SARS 2 variants,
  • 12:10at least the variants that
  • 12:11were prevalent at the time.
  • 12:13So you know,
  • 12:15this was totally unexpected
  • 12:17because what we found was that
  • 12:19this monoclonal antibody could not
  • 12:21only have this wide diverse breath
  • 12:23against these zoonotic viruses,
  • 12:25but it could also block all
  • 12:27the various variants.
  • 12:29At the time that we published a study,
  • 12:31the delta was sort of the prevalent
  • 12:32variant but it could potently
  • 12:34neutralize these various variants
  • 12:37at I see 50 levels in the sub
  • 12:39100 microgram levels.
  • 12:41And then finally in collaboration with
  • 12:43Primavera Chara at Duke University
  • 12:45who's a structural biologist.
  • 12:47Then we solve the structure for
  • 12:52DH1047 bound to the source Kobe 1RB.
  • 12:54DI don't have time to tell you,
  • 12:55tell you all about the details,
  • 12:57but what I want to just briefly
  • 12:59tell you is that the DH1047,
  • 13:01this broadly neutralizing and broadly
  • 13:03protective antibody against our
  • 13:05Becca viruses then actually binds to
  • 13:07the inner side of this or Becca virus RBD.
  • 13:10So in a more detailed paper that
  • 13:11was published by Eric Holman,
  • 13:13Sapphire and colleagues then an
  • 13:15antibody that they described called
  • 13:17RB D6 or COVID 250 then actually has
  • 13:20almost the identical footprint of this
  • 13:22particular antibody that I told you.
  • 13:25So not only did we demonstrate that a
  • 13:28DH1047 has his broad activity in mice,
  • 13:29but on a more practical level,
  • 13:31we think that the epitope or the
  • 13:34footprint bound by this antibody
  • 13:35is actually a rational target
  • 13:38for panzerbecovirus vaccines.
  • 13:39And we're actually interested in utilizing
  • 13:41this knowledge for deploying and
  • 13:43developing optimal vaccination strategies
  • 13:45against this group of SARS related viruses.
  • 13:50In fact, that some of our
  • 13:51work also spans this area.
  • 13:53So I think it's also clear that no
  • 13:56universal vaccines exist against
  • 13:58this group of SARS related viruses,
  • 14:00although we have a number of
  • 14:02individuals that are working on
  • 14:04approaches that have so far proved
  • 14:05to be successful in animal models.
  • 14:08So we designed a vaccine approach
  • 14:10that we were interested in really
  • 14:12based on a heterologous boost to
  • 14:13try to harness the immune system to
  • 14:16target these cross reactive B cells
  • 14:18to these conserve sites including
  • 14:20the site that is targeted by DH1047
  • 14:22or even more conserve sites within
  • 14:24the machinery of the S2 domain.
  • 14:26So the subunit 2 or the stock
  • 14:28that is shown here in green.
  • 14:30So some of these conserve sites include
  • 14:33the fusion peptide as well as the
  • 14:35stem Helix loop that have now been
  • 14:38described by David Weissler and colleagues.
  • 14:40And these sites can actually are
  • 14:42highly conserved among these viruses.
  • 14:44And not just within these viruses,
  • 14:46but the fusion peptide is sort of a,
  • 14:47you know, this,
  • 14:49you know,
  • 14:51general type of protein motif on
  • 14:55type 1 membranes of these various
  • 14:58infectious viruses that infect humans.
  • 15:01So I don't have time to tell you
  • 15:03all about the details of this data.
  • 15:04But utilizing the design of
  • 15:06these chimeric spikes,
  • 15:08then we aim to boost mice in a manner
  • 15:10that could direct their immune systems
  • 15:12to these highly conserved areas.
  • 15:15And we aim to try to increase
  • 15:17immune coverage against these
  • 15:18genetically divergent Sarbeca
  • 15:19viruses that are shown here on the
  • 15:22left by this phylogenetic tree.
  • 15:23So just to tell you a little
  • 15:25bit of the details of this data.
  • 15:26Then the chimeric spike boosted mice
  • 15:28that I I am telling you about are
  • 15:32actually shown in Group one or group 2.
  • 15:35So they were either given to the mice in
  • 15:37a Multiplex manner or boosted separately.
  • 15:40And the what I want to draw your
  • 15:43attention to is that Group 4 is
  • 15:46actually AM RNA vaccinated mouse
  • 15:48group that received a similar vaccine
  • 15:50to the Pfizer and Moderna vaccine.
  • 15:52So whenever we vaccinate these mice
  • 15:54and begin to compare the neutralizing
  • 15:56activities of these various
  • 15:58vaccines including our vaccines,
  • 16:00then what we observed with our
  • 16:01vaccines is that we were able to
  • 16:04elicit broadly neutralizing antibodies
  • 16:05not just against the original SARS
  • 16:07and SARS Co V2 but also against
  • 16:10these more genetically divergent
  • 16:12preemergent SARS related viruses.
  • 16:14More importantly and I think a
  • 16:16far more stringent test is that
  • 16:18whenever we introduce
  • 16:19viruses that were not included in
  • 16:21the vaccine cocktail to these mice
  • 16:23including a heterologous WI V1 or
  • 16:25even a very hard to neutralize at
  • 16:28the time the so-called beta variant,
  • 16:30then our vaccines can fully
  • 16:32protect mice against both lower and
  • 16:35upper airway virus replication.
  • 16:36So we think that this could be a
  • 16:39potentially a strategy among many strategies.
  • 16:41So this is by no means you know that
  • 16:43one of the by no means the only approach
  • 16:46that different individuals are pursuing.
  • 16:48But we think that this could be a
  • 16:52strategy to to have a tool in our
  • 16:54armamentarium for dealing with these
  • 16:55genetically divergent viruses that
  • 16:57could emerge at a later point in time.
  • 16:59And we think that this could perhaps
  • 17:01be a strategy for preventing,
  • 17:03God forbid,
  • 17:04a future starts Kobe 3 virus emergence event.
  • 17:07So this is clearly one of the
  • 17:09approaches that we think is successful.
  • 17:11But there are other approaches
  • 17:13that not only us,
  • 17:14but others are pursuing in the field.
  • 17:16And one of the approaches that I
  • 17:17want to sort of end with in terms of
  • 17:20the coronavirus aspect of my talk is
  • 17:22to tell you a little bit about how
  • 17:24we're thinking about dealing with
  • 17:26more of the breadth of these various
  • 17:29highly pathogenic coronaviruses.
  • 17:30So in a collaboration with Duke University,
  • 17:33then what we began to do is to engineer
  • 17:37nanoparticles by including various RBD's.
  • 17:39So the RBD remember is a site that
  • 17:41contains these Achilles heels on
  • 17:43these various viruses and one of the
  • 17:45areas that we're interested in is to
  • 17:47be able to expand the immune breath,
  • 17:49the protective immune breath
  • 17:51against these various viruses.
  • 17:52So in a proof of concept study that I'm
  • 17:55going to just tell you about real quick,
  • 17:57then what we decided to do was to expand
  • 18:00the breath of our our vaccine that
  • 18:03I'm going to tell you about not just
  • 18:05against group to be Cerbecca viruses,
  • 18:07but also group,
  • 18:08group to see Marbecca viruses.
  • 18:10So we engineered A ferritin nanoparticle
  • 18:14vaccine and tagged three of these
  • 18:16various receptor binding domains
  • 18:18from not only group to obese,
  • 18:19but also group to C sarbacoviruses.
  • 18:22And we began to study the immunogenicity
  • 18:24and the ability of the epitopes on
  • 18:26the receptor binding domains to
  • 18:28be presented in a manner that the
  • 18:30immune system immune system could
  • 18:32see them and generate optimal immune
  • 18:34responses in at least in mouse models.
  • 18:36So I don't have time to tell you
  • 18:38all about the details of that data,
  • 18:39but I'll show you some of the data
  • 18:42that we think is important and and
  • 18:44being able to guide universal vaccine
  • 18:47approaches.
  • 18:47So the first question that we asked
  • 18:49is whether or not this multimeric
  • 18:51nanoparticle RBD vaccine could elicit
  • 18:53this protective immunity against
  • 18:54these genetically divergent not
  • 18:56just SARS related virus,
  • 18:58but also MERS related viruses that
  • 19:00we know are poised for emergence
  • 19:03since MERS coronavirus continues to
  • 19:05emerge each year in the Middle East.
  • 19:07But we wanted to test these viruses,
  • 19:10test these this vaccine against
  • 19:12these viruses not just in stringent
  • 19:15in vitro assays but also in highly
  • 19:18pathogenic animal models of of disease.
  • 19:21So when we evaluated the ability of these
  • 19:23of the vaccine to elicit protective
  • 19:26immunity in particular neutralizing
  • 19:27antibodies against authentic viruses.
  • 19:30So this this is not pseudovirus
  • 19:32but actually authentic virus.
  • 19:34Then this Multimeric RBD vaccine not only
  • 19:37elicited high levels of neutralizing
  • 19:40antibodies against zoonotic viruses,
  • 19:43also the original sarscovy from 2003,
  • 19:46but we could also elicit whenever
  • 19:49we begin to add more relevant RBD's
  • 19:53neutralizing antibodies against
  • 19:55this other group to see MERS, MERS,
  • 19:58coronavirus as well as a whole
  • 19:59host of other MERS related viruses.
  • 20:02Which I don't have time to tell you
  • 20:04about that data and a more stringent
  • 20:06test whenever we evaluate the vaccine
  • 20:09and not just protecting against
  • 20:11mortality and a highly pathogenic
  • 20:13SARS Co V1 mouse model,
  • 20:14but what we what we see is that.
  • 20:17The multimeric vaccine can not
  • 20:19only protect against lung virus
  • 20:21replication relative to a control,
  • 20:23but it can also protect against
  • 20:26upper airway virus replication.
  • 20:27And then similarly whenever we evaluate
  • 20:29the ability of this trivalent or
  • 20:32multimeric vaccine to protect against
  • 20:34MERS coronavirus then we see fairly
  • 20:36stringent protection against both lower
  • 20:38and upper airway virus replication.
  • 20:41So I'm in conclusion then this is another
  • 20:44approach that we're excited about.
  • 20:45So the so-called multimeric vaccine
  • 20:48expressing various Group 2B and
  • 20:50Group 2C receptor binding domains
  • 20:52to elicit these types of broadly
  • 20:54neutralizing antibodies.
  • 20:55And we think that this could be one
  • 20:58of the many strategies that not only
  • 21:00us but in other individuals that
  • 21:02are pursuing similar strategies.
  • 21:03In particular,
  • 21:04I'm David Viessler from the University
  • 21:06of Washington and Howard Hughes
  • 21:08Medical Institute as well as Pamela
  • 21:10Bjorkman who also have other really
  • 21:12promising approaches.
  • 21:12So I would be remiss not to
  • 21:14mention their work in this area,
  • 21:16which I think is also critically
  • 21:20important and some future directions,
  • 21:21at least for this particular part of my talk.
  • 21:24Then you know,
  • 21:25we're of course interested in
  • 21:26evaluating whether or not we can
  • 21:28begin to boost some of these conserved
  • 21:30epitopes within the S2 machinery.
  • 21:32So in particular the fusion peptide
  • 21:33as well as the same Helix loop,
  • 21:35irrelevant animal models,
  • 21:37as well as these more multimeric vaccines.
  • 21:40And not just their ability to
  • 21:42prevent mortality and highly
  • 21:43pathogenic disease in mice,
  • 21:45but also the ability to prevent
  • 21:47the transmission of these highly
  • 21:49transmissible SARS Cov 2 variants.
  • 21:52Another area that I'm actually
  • 21:53just super interested in and will
  • 21:55be an area that the lab will be
  • 21:58expanding into is understanding
  • 22:00sort of more fundamental mechanisms
  • 22:02of the induction and generation
  • 22:05and maintenance of plasma cells.
  • 22:07And in particular long lived
  • 22:08plasma cells and relevant tissues.
  • 22:10Not just in the bone marrow,
  • 22:11but also in relevant areas including
  • 22:14the lemon appropriate or even
  • 22:17in the upper and lower airway.
  • 22:19Utilizing this really nice and
  • 22:22elegant system for time stamping
  • 22:24and being able to track the
  • 22:28generation and longevity of
  • 22:29these cells in various tissues.
  • 22:31So in the last couple minutes then
  • 22:33I just want to just briefly tell
  • 22:35you about another group of viruses.
  • 22:36So I I mentioned very early
  • 22:38in my talk about, you know,
  • 22:39the 26 viral families that can infect humans.
  • 22:42So Corona Verde is one of those
  • 22:43families that I just told you about.
  • 22:45But another of those families
  • 22:47includes the Flavy Verde,
  • 22:48which as we also know has been a a major,
  • 22:54you know, source of human infections.
  • 22:58And in particular,
  • 22:59including members from from from
  • 23:01this group have made many important
  • 23:04contributions whenever Zika virus which
  • 23:06is a flaming virus emerged in 2015.
  • 23:09But a virus that I'm actually really
  • 23:11interested in is dengue virus.
  • 23:13And the reason why I'm interested in
  • 23:15this virus because it actually breaks
  • 23:18the many of the canonical immunological
  • 23:20rules that we typically think of in
  • 23:23terms of understanding antibody responses.
  • 23:25So this virus is transmitted
  • 23:27by a many vectors.
  • 23:29One of these vectors includes 80S aegypti
  • 23:32and it's highly prevalent in tropical
  • 23:34areas including my very own El Salvador.
  • 23:37And it infects upwards of
  • 23:39400 million people each year.
  • 23:41And the group that's most afflicted
  • 23:43by the most severe forms of
  • 23:46the disease includes children.
  • 23:48And one of the areas that I'm interested
  • 23:50in studying Danga virus is really
  • 23:52more on the immune side of things.
  • 23:54So and now a published paper as well
  • 23:58as other epidemiologic papers in
  • 24:01the New England Journal of Medicine.
  • 24:03Then it's become increasingly clear
  • 24:06that in zero negative children that
  • 24:09received the only FDA approved vaccine,
  • 24:11the socalled Invaxia,
  • 24:12then these children have higher
  • 24:15incidence of hospitalization in very
  • 24:17young children compared to children who
  • 24:20have had a prior dengue virus infection.
  • 24:23So if you have,
  • 24:23if you're a zero negative child,
  • 24:25you live in an endemic area,
  • 24:27you cannot actually get the vaccine.
  • 24:29It's actually contraindicated
  • 24:30in this particular group.
  • 24:32So Aravinda De Silva is one of my
  • 24:36postdoc mentors and he's actually
  • 24:39supporting my program by letting me
  • 24:42access PBMC samples from a cohort from
  • 24:44the Philippines that which contains 0
  • 24:47negative and 0 positive children who
  • 24:49were vaccinated with being vaccia.
  • 24:50So I'm actually really interested
  • 24:53in understanding the fundamental
  • 24:54properties of those monoclonal
  • 24:56antibodies from these by basic groups
  • 24:58of of children in terms of their
  • 25:01baseline status and understanding what
  • 25:03is particular about these antibody
  • 25:05properties and these children that
  • 25:07could potentially lead to enhanced disease.
  • 25:10So just again,
  • 25:12not really give you everything,
  • 25:13but just to give you a little bit
  • 25:15of the flavor of the things that
  • 25:16we're interested in for the future.
  • 25:18So with that,
  • 25:19I'd like to acknowledge members who
  • 25:21contributed to the to these studies,
  • 25:22including members from UNC particular,
  • 25:26my postdoctoral advisor Ralph Barrick
  • 25:28and Aravinda De Silva from Duke University,
  • 25:32Bart Haynes,
  • 25:32Kevin Saunders.
  • 25:33And then our collaborator on all
  • 25:36the mRNA work,
  • 25:37Drew Wiseman and also a nascent
  • 25:40collaborator who's also assistant
  • 25:42professor at University of
  • 25:44Pennsylvania Norbert Party,
  • 25:46who did his post for Drew.
  • 25:48We're also going to be collaborating
  • 25:50with him on on mRNA approaches
  • 25:51to emerging flaving viruses.
  • 25:53So more to come on that.
  • 25:55Also my funding acknowledgements from
  • 25:57the Howard Hughes Medical Institute.
  • 25:59Thank you very much for your attention. So
  • 26:11you're good with time. So we can take
  • 26:13some questions from the audience.
  • 26:23So I have the microphone.
  • 26:31Excellent talk. Thank you, David.
  • 26:33My question is about the
  • 26:35conserved epitope to you.
  • 26:37You found that the monocle
  • 26:39Ronnie body binds to has that.
  • 26:41How is it resistant to evasion
  • 26:44and is that there's some kind
  • 26:46of a critical function that
  • 26:47the virus has to preserve?
  • 26:49And is it also preserved
  • 26:50in the Omicron linear?
  • 26:51Yeah. So if if you incubate the
  • 26:54antibody with Omicron and this
  • 26:57various lineages for longer,
  • 26:58you can have some neutralization.
  • 27:00So even though many of the Omicron
  • 27:04lineages haven't necessarily mutated
  • 27:06the footprint of the antibody,
  • 27:07then additional mutations within the
  • 27:09RBD that are distal to the actual
  • 27:12footprint epitope can actually alter
  • 27:14the accessibility of the epitope.
  • 27:16So you know much like many of the
  • 27:20monoclonal antibody therapies then
  • 27:23Omicron and its various lineages
  • 27:25including you know the latest XBB
  • 27:271.5 and its various lineages then
  • 27:29have demonstrated the ability
  • 27:31to evade this DH1047 antibody.
  • 27:34So that's unfortunately,
  • 27:35you know, just a a,
  • 27:37a problem that the field has and and
  • 27:42something that the virus has figured out by,
  • 27:44you know,
  • 27:45mutating sort of these distal and
  • 27:47outside parts within the footprint,
  • 27:49not even necessarily the footprint
  • 27:52but but you know we're still excited
  • 27:54about the ability of this to,
  • 27:57you know,
  • 27:57retain its function against these number
  • 28:00of different zoonotic viruses that
  • 28:02we know are poised for human emergence.
  • 28:05David. Hey David, great talk.
  • 28:06Thank you.
  • 28:07I'm
  • 28:07curious what your your choice of
  • 28:10nanoparticles and is it important for the
  • 28:12these multivalent vaccines
  • 28:13for them to actually
  • 28:14be attached to a a particle or
  • 28:17that's is that advantageous compared
  • 28:19to just having mix of proteins injected.
  • 28:22Yeah, you know we think it we we
  • 28:24think it might be and you know this
  • 28:26this is certainly on on you know
  • 28:28the more practical side is this is
  • 28:29reflected by the by the potency and
  • 28:31the amount of immune response we see.
  • 28:33In particular you know the levels
  • 28:36of serum neutralizing antibodies are
  • 28:39greatly exceeded whenever we couple
  • 28:42you know whatever protein of choice
  • 28:44in this case that receptor binding
  • 28:46domain in a multi in a multimaric form.
  • 28:49You know for probably for reasons
  • 28:51of you know activating you know
  • 28:54besyl receptors in a in a better way
  • 28:57potentially or in a more potent way.
  • 28:59But that seems to be a feature that confers,
  • 29:03you know, these B cells to to make far
  • 29:06more potent levels of antibody responses.
  • 29:09And this is not something that
  • 29:10we just see with coronaviruses,
  • 29:12but also with other viruses that are
  • 29:15genetically divergent like HIV for example.
  • 29:17So let me just quick follow up.
  • 29:19That's really interesting.
  • 29:20Is it important that individual
  • 29:22particles be multivalent or if
  • 29:25you took a mixture of particles,
  • 29:281 coated with this receptor binding domain,
  • 29:31another particle coded with that one,
  • 29:34mix them together and then inject it,
  • 29:35would that work equally well? Yeah.
  • 29:37So that's that's a great question.
  • 29:38So you know based on data that from
  • 29:42from our group actually you know with
  • 29:45the with the chimeric mRNA vaccines
  • 29:47when you give them all together
  • 29:50even though you may have breath you
  • 29:52actually lose some of the potency.
  • 29:53You know and you know we could go
  • 29:55on about you know like theoretical
  • 29:57reasons underlying this actual you
  • 29:59know phenotype you know but it
  • 30:01could just be that you just have
  • 30:03you know multiple different clones
  • 30:04of B cells that are being selected
  • 30:06against the various antigens as
  • 30:08opposed to you know more potent and
  • 30:11higher affinity clones against the
  • 30:13the the single particle with say
  • 30:15like a single protein for example.
  • 30:18But we we definitely lose some of
  • 30:20the potency when we include more
  • 30:21antigens and and give them all at once.
  • 30:25So but but but definitely I that's an
  • 30:27interesting question and something
  • 30:28that we've thought about a lot
  • 30:30in optimizing these strategies.
  • 30:31Thank you.
  • 30:32That's a great talk and great discussion.
  • 30:34I am fascinated by these same topics.
  • 30:36So we'll be sure to continue
  • 30:38over the lunch break asking you.