Robust Mouse Rejuvenation

Study 1

Overview

LEV Foundation's flagship research program is a sequence of large mouse lifespan studies, each involving the administration of (various subsets of) at least four interventions that have, individually, shown promise in others' hands in extending mean and maximum mouse lifespan and healthspan.

We focus on interventions that have shown efficacy when begun only after the mice have reached half their typical life expectancy, and mostly on those that specifically repair some category of accumulating, eventually pathogenic, molecular or cellular damage. The first study in this program began in early 2023.

Related Content

Progress Report Video

Goals and Motivations

Our ultimate goal in this program is to achieve "Robust Mouse Rejuvenation". We define this as an intervention, almost certainly multi-component, that:

  • is applied to mice of a strain with a historic mean lifespan of at least 30 months

  • is initiated at an age of at least 18 months

  • increases both mean and maximum lifespan by at least 12 months

In each study in this program, we will examine the synergy of (typically at least four) interventions already known individually to extend mouse lifespan when started in mid-life.

We will determine not only the ultimate readout of lifespan, but also the interactions between the various interventions, as revealed by the differences between the treatment groups (receiving different subsets of the interventions) in respect of the trajectories with age of cause of death, decline in different functions, etc. In this way we will add greatly to the understanding of which benefits these interventions confer and how they synergize, or possibly antagonize.

There are two key motivations for this program. One is purely scientific: as with all mouse work with a biomedical end goal, we hope to generate data that will inform the development of therapies to let humans live longer in good health. The other could be called rhetorical, societal, political - it is to demonstrate a definitive proof of concept that aging is much more malleable than society currently insists on thinking it is, and thus must be viewed as a tractable medical problem, rather than a fact of life.

Study Design

Provider: Studies will be conducted at Ichor Life Sciences (Syracuse, NY). Ichor has a long-standing reputation conducting animal studies for pharma and the biotech industry, and has experience performing lifespan studies in mice. They are well-equipped for a study of this size and we know them well.

Mouse Strain: We have selected pre-aged C57Bl/6J for the first study on the basis of availability, and in order to match other lifespan studies involving the interventions employed here. The second study in the RMR program (examining a different panel of interventions) will probably be conducted in HET3 mice; this strain may be a more representative model of human aging, and will provide a valuable direct comparison to data from the NIA Interventions Testing Program. Subject to funding, we expect to be able to source a sufficient population of pre-aged HET3 mice to initiate the second study from Jackson Labs by Q3 2023.

Age: Interventions will begin at 18 months in order to assess the repair/rejuvenation capacity of interventions.

Interventions: Interventions are chosen on the basis that they 1) act systemically and 2) have individually shown some lifespan-extending effect in naturally aged mice. In this way, we are specifically selecting rejuvenation therapeutics, as opposed to those which are purely preventative and/or require early life intervention. Therapies are also selected to have minimal mechanistic overlap, based on our current understanding of their mechanisms of action.

Controls: We are using two types of control for each intervention. One is “mock”: a treatment that resembles the real treatment as closely as possible except that the “active ingredient” is absent; and one (“naive”) is the absence of any treatment. This is so that we can distinguish the impact of the treatment itself from that of the mode by which it is administered,

Treatment Groups: 500 male and 500 female mice divided into 10 groups, as follows:

  • ○     25 male, 25 female receive mocks

    ○     25 male, 25 female receive nothing

  • ○     50 male, 50 female receive rapamycin

    ○     Half of these receive mocks for the other interventions

    ○     Half of these receive rapamycin only, no mocks

  • ○     50 male, 50 female receive senolytic

    ○     Half of these receive mocks for the other interventions

    ○     Half of these receive senolytic only, no mocks

  • ○     50 male, 50 female receive mTERT

    ○     Half of these receive mocks for the other interventions

    ○     Half of these receive mTERT only, no mocks

  • ○     50 male, 50 female receive HSCT

    ○     Half of these receive mocks for the other interventions

    ○     Half of these receive HSCT only, no mocks

  • ○     50 male, 50 female receive all interventions EXCEPT rapamycin

    ○     Half of these receive empty thermoplastic in chow

    ○     Half of these have chow without thermoplastic

  • ○     50 male, 50 female receive all interventions EXCEPT senolytic

    ○     Half of these receive mock senolytic

    ○     Half of these receive no mock senolytic 

  • ○     50 male, 50 female receive all interventions EXCEPT mTERT

    ○     Half of these receive scrambled vector

    ○     Half of these receive no vector

  • ○     50 male, 50 female receive all interventions EXCEPT HSCT

    ○     Half of these undergo mobilization without HSCT

    ○     Half of these do not undergo mobilization

  • ○     50 male, 50 female mice receive all interventions

Longitudinal Assessment: All living animals will undergo functional and behavioral assessment at timepoints linked to % group survival (see Endpoints below). These assessments have been selected on the basis of being minimally invasive and stressful, while simultaneously allowing evaluation of parameters reflecting functional capacity in mice.

They will include rotarod and grip strength assessments for motor and muscle function, novel object recognition for cognition, and software-enhanced open field testing to measure movement velocity and duration, gait abnormalities, etc. as well as provide indications of openness vs anxiety. Frailty will also be scored on the basis of body condition, weight, and factors such as degree of kyphosis and alopecia on an ongoing basis.

In addition, blood samples will be taken longitudinally from all living mice for glucose tolerance testing, platelet counts, and to screen for HSCT engraftment (see Interventions). A fraction of each sample will be banked for retrospective analysis of the longest-lived mice.

Cull schedule: We will sacrifice 12 mice out of each group of 50 (males or females, for each of the ten treatments) for analyses that require terminally invasive tissue samples. In contrast to most studies, we will schedule these based not on chronological age but on group-specific survival curves, as shown in the figure. We believe that this will be more informative than the traditional approach, since the underlying correlation between biological and chronological age is factored out.

Endpoints: Male and female mice of each intervention group will be sacrificed on a declining schedule based on in-group survival (see culling schedule). An equal number of animals will be randomly selected from those receiving mock or naive control interventions (if any) for a given intervention group.

At these timepoints necropsy will be performed, along with CBC with leukocytes (to assess immune status), blood chemistry, and storage of all major organs and tissues. Downstream analyses will include measurements of serum cytokines/chemokines (inflammatory state), telomere length, and epigenetic age estimation, in addition to tissue histology.

The same intensive post-mortem analysis will also be carried out on every 4th mouse that is euthanized for humane reasons, while tissue and biofluid storage will be conducted on all expired animals where the time of death permits the collection of valid samples.

Interventions

  • Rationale: Rapamycin has been selected as our baseline intervention due to the many studies demonstrating a lifespan effect in mice, including when started late in life. mTOR inhibition by rapamycin is believed to delay the onset of cancer, an effect which may be potentiated by combination with other therapeutics.

    Design: Mice in treatment groups will receive 42ppm Eudragit S100 enteric-coated rapamycin in chow (Purina 5LG6), using the same encapsulation provider and formulary as ITP studies. Food will not be irradiated.  We have selected oral delivery in chow over other delivery methods in order to be minimally invasive and so that the drug can be administered continuously.

    A dose of 42ppm has been chosen on the basis of sex-specific dose effects [PMID 33145977], particularly the observation that male mice receive minimal or no benefit from lower doses of 14ppm [PMID 24341993], and higher doses have not shown any detrimental effects.

    Half of the mice NOT receiving rapamycin will receive chow containing empty Eudragit, while the other half will have standard chow.

  • Rationale: Senescent cells (SnC) are shown to accumulate with age in nearly all tissues, and multiple studies have now shown improvement in healthspan parameters upon SnC removal. Although few of these studies emphasize lifespan effects, we hypothesize that SnC removal is important for mouse longevity due to the role played in immune decline and in cancer – the two leading causes of death in C57Bl/6J mice. We believe an effective senolytic may not only reduce cancer incidence by removal of cells which are cancer-capable (via senescence escape), but also by improving local immune surveillance against abnormal cells, by reducing the SASP’s cloaking effect and relieving systemic immune fatigue from SASP-driven chronic inflammation. In this way, it is also possible that SnC removal can reduce the age-related increase in susceptibility to pathogens.

    Design: Discovery of effective senotherapeutic compounds remains an active area of research, with many promising drug candidates in development alongside approaches such as senolytic vaccination using CAR-T cells or conditional suicide gene therapy. Though these second-generation senolytic therapies likely have much to offer in terms of improved specificity and SnC subtype targeting, we felt the current study would benefit most from the extensive data already collected using first generation drugs, and thus selected the broad-spectrum senolytic Navitoclax for inclusion in RMR1. While navitoclax is generally effective as a pan-senolytic compound, ablating SnCs in multiple tissue types while sparing most normal calls, platelets are particularly sensitive to its mechanism of action, Bcl-xl inhibition. To minimize this undesirable side effect, we employed a galactose conjugation prodrug strategy previously developed by colleagues in the field, wherein a galactose moiety attached to the Navitoclcax molecule renders the drug inactive until it can be cleaved by lysosomal beta-galactosidase, an enzyme consistently and considerably elevated in SnCs. ‘Off-target’ activity, for example, against platelets, is greatly reduced in this way, while efficacy against target SnC populations remains largely intact.

  • Rationale: Though we recognize that telomerase gene therapy may be neither practical nor especially beneficial for humans in the near term, it has been shown to significantly extend the lifespan of naturally-aged mice without increasing cancer incidence [PMID 22585399], while also improving numerous measures of healthspan. This work, originally conducted using AAV-mTERT gene therapy, has been replicated in numerous genetic and disease models  [PMID 31624261, 27252083, 29378675], and recently was adapted as a CMV-based gene therapy [PMID 35537048], enabling repeat dosing in rodents and humans.

    Design: We will utilize an AAV-mTERT gene expression vector, administered monthly to mice from the age of 18 months. As in the recent work conducted by Church, Parrish et al. [PMID 35537048], we will utilize intranasal delivery, which the authors demonstrated to be equally beneficial versus IV administration and which allows us to limit unnecessarily invasive procedures.

    Furthermore, the model study stopped treatments upon death of all the control mice ~29 months, then resumed treatments at 32 months given the excellent health demonstrated by treatment groups. Despite this pause, aged mice that resumed mTERT therapy lived to an average 37.5 months, a 41.4% increase over controls. We have therefore chosen to continue mTERT therapy on a monthly schedule throughout the remaining lifespan of the intervention groups.

  • Rationale: A reduced regenerative capacity of stem cells is widely believed to contribute to age-related morbidity and functional tissue decline. Numerous studies have shown stem cell transplantation to have rejuvenating effects in mouse tissues, and several additionally show a lifespan extension effect [PMID 32012439, 31031800].

    Design: The study design is largely (but see below) based on the protocol utilized by Guderyon et al. 2020 [PMID 32012439] and consists of mobilizing the recipient bone marrow niche followed by transplant of lineage-depleted hematopoietic stem cells (HSCT).

    Bone Marrow: We have opted to use lineage-depleted bone marrow HSCs as opposed to additional selection for and expansion of long-term self-renewing HSCs. This was on the basis that 1) prior lifespan studies used whole or lineage-depleted bone marrow, which may include MSCs or other beneficial cells promoting engraftment, and 2) expansion protocols have not been extensively validated.

    Mobilization Protocol: We will follow the mobilization protocol for recipient mice as outlined in Guderyon 2020, consisting of G-CSF + AMD3100. Although other newer mobilization reagents require fewer injections, efficacy is not well established; the current protocol is most common for chemical mobilization, has been used in aged mice, and remains the current standard of care for mobilizing HSCs from human BM donors.

    # of cells: Each administration will consist of ~2x10⁷ HSCs, derived from 8 sex-matched C57Bl/6J  PEP-BOY (CD45.2) donor mice per injection. This number is four-fold greater than the Guderyon study, however in line with other HSCT studies [PMID 31031800, 18491294]. Additionally, it is demonstrated that % donor cell engraftment is largely based on competition with mobilized recipient cells; thus, increasing the donor to recipient ratio in the blood may allow for increased engraftment with each administration allowing for fewer transplants overall and thus less stress on the animals.

    Number of Transplant Injections: While the lifespan cohort of the Guderyon study received a total of 8 HSCTs, each round administered only 5x10⁶ lineage-negative (long-term repopulating) donor cells. We wish to minimize the number of administrations for each mouse. Therefore, we will perform 2 rounds of HSCT, a month apart, and then assess the % donor-derived cells (CD45.2) in peripheral blood after 4-8 weeks (timing to coincide with planned blood draw). Whether or not additional rounds of HSCT are performed (for a given experimental group) will be decided based on percentage engraftment in recipient mice.

Future Interventions

As of November 2023, high-level design of the second study in the RMR program is largely complete. A summary of our plans for RMR2 is now available.