Someone from the lab or who worked on the paper almost always shares the full access paper on twitter… here it is:

3 Likes

This still comes out paywalled for me; do you get the full text from that link?

This twitter post has the full paper link (not sure why it doesn’t work if I copy the article link from the twitter post):

https://twitter.com/JPCastro_Aging/status/1664887049122349057

1-s2.0-S0092867423004762-mainsm.pdf (2.2 MB)

2 Likes

They started treatment only at 25 months, with average lifespan around 30 months, and still got 11% median life extension and 33% remaining median life extension. Also max lifespan in the treatment group is ~150 days longer than in control. Pretty impressive! How does it compare to rapamycin?

2 Likes

Below are some good snippets. The paper is currently in press, so the figures are low quality at this point. I’m sure they will be better once it’s published.

Has anyone looked into ascorbyl-palmitate? Seems to be available as a supplement.

Since chronic rapamycin treatment also leads to glucose intolerance in mice,[131] we we performed a glucose tolerance test on a separate cohort of 24-month-old male mice subjected to KU0063794 for 2 months. KU0063794 did not affect glucose tolerance in old mice as there was no difference in glucose clearance dynamics between the control and treated groups (Figures 7H and 7I).

By comparing expression profiles of treated mice with age matched control samples, we identified compound-induced ECs in murine organs and examined their association with biomarkers of aging and longevity (Table S6B). Consistent with CMap predictions, all selected compounds generated changes positively associated with an aggregated signature of at least one longevity model (Figure 7A, lower). Moreover, pro-longevity effects of KU0063794, ascorbyl-palmitate, AZD8055, and GDC0941 were supported simultaneously by aggregated biomarkers of lifespan-extending interventions (in the kidney and the liver; p adjusted < 0.004) and long-lived species (in kidney; p adjusted < 6 3 10"4 ), along with multiple individual signatures (GH deficiency, CR, maximum and median lifespan, etc.). To test if compounds that induce longevity-associated ECs extend murine lifespan and healthspan, we subjected 25-month-old C57BL/6 male mice to a diet containing a top hit from our analysis, KU0063794 (Figure 7B). KU0063794 at 10 ppm extended the remaining median and ML of old mice by 32.6% and 10.9%, respectively (log-rank test p = 0.038) (Figure 7C), with no effect on animal body weight (Figure 7D). KU0063794 also improved mouse gait speed measured at 30 months (Figure 7E). The frailty index of mice before and 5 months after the treatment initiation showed no difference between the control and experimental groups prior to drug supplementation (Figure S7A); however, mice subjected to KU0063794 were significantly less frail following the treatment (Figure 7F). Detailed analyses also revealed a KU0063794- induced improvement of coat and eye-related features (Figure S7B).

4 Likes

Do we have any chemists here?

MATERIALS AND METHODS

Materials

Protein G–Sepharose and glutathione–Sepharose were purchased from Amersham Bioscience. [γ-32P]ATP was from PerkinElmer; IGF1 (insulin-like growth factor) was from Invitrogen. Tween 20, DMSO, PMA and dimethyl pimelimidate were from Sigma, and CHAPS and rapamycin were from Calbiochem. Akti-1/2, PI-103 and PD0325901-CL were synthesized by Dr Natalia Shpiro at the MRC Protein Phosphorylation Unit, University of Dundee. Ku-0063794 was synthesized at AstraZeneca. The wild-type control mLST8+/+ and mLST8−/− knockout MEFs (mouse embryonic fibroblasts) were described previously [17] and provided by David Sabatini (Whitehead Institute for Biomedical Research, Cambridge, MA, U.S.A.). The wild-type control Rictor+/+ and Rictor−/− knockout MEFs were described previously [29] and provided by Mark Magnuson (Vanderbilt University School of Medicine, Nashville, TN, U.S.A.). The wild-type control Sin1+/+ and Sin1−/− knockout MEFs were described previously [16] and provided by Bing Su (Yale University School of Medicine, New Haven, CT, U.S.A.).

I’ve been taking it for a few years now… research looks positive, no obvious downsides:

See on Amazon:
https://www.amazon.com/gp/product/B001F0R66A/ref=ppx_yo_dt_b_search_asin_title?ie=UTF8&psc=1

Also - some interesting information on the other compounds mentioned:

AZD8055

The serine/threonine kinase mammalian target of rapamycin (mTOR) is crucial for cell growth and proliferation, and is constitutively activated in primary acute myeloid leukemia (AML) cells, therefore representing a major target for drug development in this disease. We show here that the specific mTOR kinase inhibitor AZD8055 blocked mTORC1 and mTORC2 signaling in AML. Particularly, AZD8055 fully inhibited multisite eIF4E-binding protein 1 phosphorylation, subsequently blocking protein translation, which was in contrast to the effects of rapamycin. In addition, the mTORC1-dependent PI3K/Akt feedback activation was fully abrogated in AZD8055-treated AML cells. Significantly, AZD8055 decreased AML blast cell proliferation and cell cycle progression, reduced the clonogenic growth of leukemic progenitors and induced caspase-dependent apoptosis in leukemic cells but not in normal immature CD34+ cells. Interestingly, AZD8055 strongly induced autophagy, which may be either protective or cell death inducing, depending on concentration. Finally, AZD8055 markedly increased the survival of AML transplanted mice through a significant reduction of tumor growth, without apparent toxicity. Our current results strongly suggest that AZD8055 should be tested in AML patients in clinical trials.

More here:
https://www.nature.com/articles/leu2011339

Preclinical pharmacology
AZD8055 is a potent, selective inhibitor of mTOR kinase, targeting both mTORC1 (rapamycin-sensitive) and mTORC2 (rapamycin insensitive) complexes. AZD8055 is specific against mTOR (IC50 value of 0.8 ± 0.2 nM using an immunoprecipitate of full length mTOR from HeLa cells in an ELISA-based kinase assay). AZD8055 was inactive in a counter screen against 260 kinases.

AZD8055 inhibited downstream targets of both mTORC1 (phosphorylation of S6 at serine 235/236) and mTORC2 (phosphorylation of AKT at serine 473) in several in vitro models in a dose- and time-dependent manner. Oral treatment of mice bearing U87-MG human glioma xenografts twice daily with 2.5, 5, and 10 mg/kg/day AZD8055 for 10-days resulted in a dose-dependent tumour growth inhibition of 33%, 48% and 77%, respectively. All doses were well tolerated. The growth inhibitory effect of once daily administration with 10 and 20 mg/kg of AZD8055 for 10-days was 57 and 85%, respectively. This tumour growth inhibitory effect was observed in a number of xenograft models tested in vivo. Furthermore, the antitumour activity in vivo was in good agreement with growth inhibition observed in cell lines in vitro.

More here:

https://openinnovation.astrazeneca.com/preclinical-research/preclinical-molecules/azd8055.html

GDC0941 / Pictilisib

Purpose

This first-in-human dose-escalation trial evaluated the safety, tolerability, maximal tolerated dose (MTD), dose limiting toxicities (DLTs), pharmacokinetics, pharmacodynamics and preliminary clinical activity of pictilisib (GDC-0941), an oral, potent and selective inhibitor of the Class I phosphatidylinositol-3-kinases (PI3K)

Results

Pictilisib was well-tolerated. The most common toxicities were grade 1-2 nausea, rash and fatigue while the DLT was grade 3 maculopapular rash (450mg, 2 of 3 patients; 330mg, 1 of 7 patients). The pharmacokinetic profile was dose-proportional and supported once-daily dosing. Levels of phosphorylated serine-473 AKT were suppressed >90% in platelet rich plasma at 3 hours post-dose at the MTD and in tumor at pictilisib doses associated with AUC >20uM.hr. Significant increase in plasma insulin and glucose levels, and >25% decrease in 18F-FDG uptake by PET in 7 of 32 evaluable patients confirmed target modulation. A patient with V600E BRAF mutant melanoma and another with platinum-refractory epithelial ovarian cancer exhibiting PTEN loss and PIK3CA amplification demonstrated partial response by RECIST and GCIG-CA125 criteria, respectively.

Conclusion

Pictilisib was safely administered with a dose-proportional pharmacokinetic profile, on-target pharmacodynamic activity at dose levels ≥100mg and signs of antitumor activity. The recommended Phase II dose was continuous dosing at 330mg once-daily.

A Phase 2 trial with another drug/combination:

Conclusion

Adding pictilisib to anastrozole significantly increases suppression of tumor cell proliferation in luminal B primary breast cancer.

Phase II Randomized Preoperative Window-of-Opportunity Study of the PI3K Inhibitor Pictilisib Plus Anastrozole Compared With Anastrozole Alone in Patients With Estrogen Receptor–Positive Breast Cancer

https://ascopubs.org/doi/10.1200/jco.2015.63.9179

Availabilty and Pricing:

3 Likes

Excessive saturated free fatty acids (SFFAs; e.g. palmitate) in blood are a pathogenic factor in diabetes, obesity, cardiovascular disease and liver failure. In contrast, monounsaturated free fatty acids (e.g. oleate) prevent the toxic effect of SFFAs in various types of cells. The mechanism is poorly understood and involvement of the mTOR complex is untested. In the present study, we demonstrate that oleate preconditioning, as well as coincubation, completely prevented palmitate-induced markers of inflammatory signaling, insulin resistance and cytotoxicity in C2C12 myotubes. We then examined the effect of palmitate and/or oleate on the mammalian target of rapamycin (mTOR) signal path and whether their link is mediated by AMP-activated protein kinase (AMPK). Palmitate decreased the phosphorylation of raptor and 4E-BP1 while increasing the phosphorylation of p70S6K. Palmitate also inhibited phosphorylation of AMPK, but did not change the phosphorylated levels of mTOR or rictor. Oleate completely prevented the palmitate-induced dysregulation of mTOR components and restored pAMPK whereas alone it produced no signaling changes. To understand this more, we show activation of AMPK by metformin also prevented palmitate-induced changes in the phosphorylations of raptor and p70S6K, confirming that the mTORC1/p70S6K signaling pathway is responsive to AMPK activity. By contrast, inhibition of AMPK phosphorylation by Compound C worsened palmitate-induced changes and correspondingly blocked the protective effect of oleate. Finally, metformin modestly attenuated palmitate-induced insulin resistance and cytotoxicity, as did oleate. Our findings indicate that palmitate activates mTORC1/p70S6K signaling by AMPK inhibition and phosphorylation of raptor. Oleate reverses these effects through a metformin-like facilitation of AMPK.

From:

Palmitate activates mTOR/p70S6K through AMPK inhibition and...

Excessive saturated free fatty acids (SFFAs; e.g. palmitate) in blood are a pathogenic factor in diabetes, obesity, cardiovascular disease and liver failure. In contrast, monounsaturated free fatty acids (e.g. oleate) prevent the toxic effect of...

It doesn’t seem that palmitate (palmitic acid) is the same (functionally) as Ascorbyl palmitate

Palmitates are the salts and esters of palmitic acid. The palmitate anion is the observed form of palmitic acid at physiologic pH (7.4). Palmitic acid is the most common SFA found in plants, animals, and many microorganisms.

1 Like

that totally crazy! I missed it

This Ku0063794 study show that we also need to inhibit MTORC2 ! AND that inhibit MTORC2 will not increase blood glucose and the usual side effect of rapamycin !

Its literally the opposite of what is believed here in this whole forum

1 Like

I think what this new study reminds us is simply that we really don’t understand mTORC2 very well yet.

Here are some research findings about mTORC2 / Rictor that suggest you don’t want to inhibit it:

From Dudley Lamming: Evidence that mTORC2 inhibition is detrimental, by Dudley Lamming

From Wikipedia:

Studies using mice with tissue-specific loss of Rictor , and thus inactive mTORC2, have found that mTORC2 plays a critical role in the regulation of glucose homeostasis. Liver-specific disruption of mTORC2 through hepatic deletion of the gene Rictor leads to glucose intolerance, hepatic insulin resistance, decreased hepatic lipogenesis, and decreased male lifespan.[46][47][48][49][50]Adipose-specific disruption of mTORC2 through deletion of Rictor may protect from a high-fat diet in young mice,[51] but results in hepatic steatosis and insulin resistance in older mice.[52] The role of mTORC2 in skeletal muscle has taken time to uncover, but genetic loss of mTORC2/Rictor in skeletal muscle results in decreased insulin-stimulated glucose uptake, and resistance to the effects of an mTOR kinase inhibitor on insulin resistance, highlighting a critical role for mTOR in the regulation of glucose homeostasis in this tissue.[53][54][55] Loss of mTORC2/Rictor in pancreatic beta cells results in reduced beta cell mass and insulin secretion, and hyperglycemia and glucose intolerance.[56] mTORC2 activity in the hypothalamus of mice increases with age, and deletion of Rictor in hypothalamic neurons promotes obesity, frailty, and shorter lifespan in mice.

Source: mTORC2 - Wikipedia

1 Like

seems not that clear here as well as they state mTORC2 activity in the hypothalamus of mice increases with age, and deletion of Rictor in hypothalamic neurons promotes obesity, frailty, and shorter lifespan in mice.

1 Like

Yes, and we don’t know whether that increase in mTORC2 activity in the hypothalamus is a good thing or a bad thing. Lots of things tend to go “up” with aging (think lipids/apoB, CRP, etc. as shown in the graph below, but increasing could be bad (i.e. an indication of damage) or good (as in damage repair).

1 Like

still this drug inhibit both and still increase lifespan in a way that ressemble rapamycin… Without the side effect…

I don’t think we know that yet - have you seen any clinical trial data in humans? All the science that I’ve seen so far suggests if you inhibit mTORC2 for a longer period of time (weeks?) then you start to get the side effects of glucose and lipid dis regulation. We’ll see if this compound somehow avoids doing this… only time and more testing will tell us this.

IMO: Unless you have preconditions, these are easily kept in check without statins or metformin if you don’t like them.

1 Like

You mean by things like bempadoic acid, acarbose, etc.?

its still too early (only mice…) but read that :

" Since chronic rapamycin treatment also leads to glucose intolerance in mice, we performed a glucose tolerance test on a separate cohort of 24-month-old male mice subjected to KU0063794 for 2 months. KU0063794 did not affect glucose tolerance in old mice as there was no difference in glucose clearance dynamics between the control and treated groups"

and we know rapamycin, both in mice and human impair glucose tolerance

1 Like

Yes, they also need to test lipid levels (mice don’t die of cardiovascular issues so a high lipid level may kill humans but not mice), and immune system function. But yes, those results with the glucose tolerance test sounds promising.

I think the most promising compound right now is the new mTOR1 selective inhibitors from Tornado Therapeutics. See more here: Joan Mannick on Rapamycin Longevity Series | Turning down mTOR to young levels may be good for aging

e: Joan Mannick, Translating Aging Podcast: “Taking disease by sTORm”: Developing Rapalogs to Extend Healthy Lifespan"

Yes. When I first started rapamycin I freaked out a little because my lipids and glucose were well in range. and then spiked when I started rapamycin.

I found out over time that things like berberine, bempadoic, acid, etc., brought back everything into a good range. The other things that have a very significant effect on glucose and lipids of course are diet and exercise.

1 Like